ArticlePDF Available

Beneficial effects of fish and fish peptides on main metabolic syndrome associated risk factors: Diabetes, obesity and lipemia

Taylor & Francis
Critical Reviews In Food Science and Nutrition
Authors:

Abstract

The definition of metabolic syndrome (MetS) fairly varies from one to another guideline and health organization. Per description of world health organization, occurrence of hyperinsulinemia or hyperglycemia in addition to two or more factors of dyslipidemia, hypoalphalipoproteinemia, hypertension and or large waist circumference factors would be defined as MetS. Conventional therapies and drugs, commonly with adverse effects, are used to treat these conditions and diseases. Nonetheless, in the recent decades scientific community has focused on the discovery of natural compounds to diminish the side effects of these medications. Among many available bioactives, biologically active peptides have notable beneficial effects on the management of diabetes, obesity, hypercholesterolemia, and hypertension. Marine inclusive of fish peptides have exerted significant bioactivities in different experimental in-vitro, in-vivo and clinical settings. This review exclusively focuses on studies from the recent decade investigating hypoglycemic, hypolipidemic, hypercholesterolemic and anti-obesogenic fish and fish peptides. Related extraction, isolation, and purification methodologies of anti-MetS fish biopeptides are reviewed herein for comparison purposes only. Moreover, performance of biopeptides in simulated gastrointestinal environment and structure-activity relationship along with absorption, distribution, metabolism, and excretion properties of selected oligopeptides have been discussed, in brief, to broaden the knowledge of readers on the design and discovery trends of anti-MetS compounds.
Full Terms & Conditions of access and use can be found at
https://www.tandfonline.com/action/journalInformation?journalCode=bfsn20
Critical Reviews in Food Science and Nutrition
ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/bfsn20
Beneficial effects of fish and fish peptides on
main metabolic syndrome associated risk factors:
Diabetes, obesity and lipemia
Soheila Abachi, Geneviève Pilon, André Marette, Laurent Bazinet & Lucie
Beaulieu
To cite this article: Soheila Abachi, Geneviève Pilon, André Marette, Laurent Bazinet & Lucie
Beaulieu (2022): Beneficial effects of fish and fish peptides on main metabolic syndrome associated
risk factors: Diabetes, obesity and lipemia, Critical Reviews in Food Science and Nutrition, DOI:
10.1080/10408398.2022.2052261
To link to this article: https://doi.org/10.1080/10408398.2022.2052261
Published online: 17 Mar 2022.
Submit your article to this journal
View related articles
View Crossmark data
REVIEW
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION
Beneficial effects of fish and fish peptides on main metabolic syndrome
associated risk factors: Diabetes, obesity and lipemia
Soheila Abachia,b, Geneviève Pilona,c, André Marettea,c, Laurent Bazineta,b,d and Lucie Beaulieua,b
aInstitute of Nutrition and Functional Foods (INAF), Université Laval, Quebec, Quebec, Canada; bDepartment of Food Science, Faculty of
Agricultural and Food Sciences, Université Laval, Quebec, Quebec, Canada; cDepartment of Medicine, Faculty of Medicine, Cardiology Axis of
the Quebec Heart and Lung Institute, Quebec, Quebec, Canada; dLaboratory of Food Processing and ElectroMembrane Processes (LTAPEM),
Université Laval, Quebec, Quebec, Canada
ABSTRACT
The definition of metabolic syndrome (MetS) fairly varies from one to another guideline and health
organization. Per description of world health organization, occurrence of hyperinsulinemia or
hyperglycemia in addition to two or more factors of dyslipidemia, hypoalphalipoproteinemia,
hypertension and or large waist circumference factors would be defined as MetS. Conventional
therapies and drugs, commonly with adverse effects, are used to treat these conditions and
diseases. Nonetheless, in the recent decades scientific community has focused on the discovery
of natural compounds to diminish the side effects of these medications. Among many available
bioactives, biologically active peptides have notable beneficial effects on the management of
diabetes, obesity, hypercholesterolemia, and hypertension. Marine inclusive of fish peptides have
exerted significant bioactivities in different experimental in-vitro, in-vivo and clinical settings. This
review exclusively focuses on studies from the recent decade investigating hypoglycemic,
hypolipidemic, hypercholesterolemic and anti-obesogenic fish and fish peptides. Related extraction,
isolation, and purification methodologies of anti-MetS fish biopeptides are reviewed herein for
comparison purposes only. Moreover, performance of biopeptides in simulated gastrointestinal
environment and structure-activity relationship along with absorption, distribution, metabolism,
and excretion properties of selected oligopeptides have been discussed, in brief, to broaden the
knowledge of readers on the design and discovery trends of anti-MetS compounds.
Introduction
Metabolic syndrome (MetS) instigated physiological, bio-
chemical, clinical, and metabolic abnormalities could seri-
ously debilitate the affected individual and contribute to
an upsurge of global mortality rates. MetS, as a cluster of
cardiometabolic risk factors, is strongly associated with
increasing predominance of various chronic conditions like
arthritis, schizophrenia including atherosclerotic cardiovas-
cular disease and cancer. According to a survey, prevalence
of MetS took a sharp leap from 1988 to 2012 among adults,
≥ 18 years of age, totaling one in three of the entire US
population. Two-thirds of US population are overweight
and or obese thus obesity, according to Yang et al., is at
epidemic proportions which should directly be confronted
to prevent MetS outbreak (Moore, Chaudhary, and
Akinyemiju 2017). Obesity, also heritable, is strongly asso-
ciated with increased morbidity and mortality. Cardiac
fatality is 40-times more likely to occur in the obese rather
than the non-obese population (Messerli et al. 1987).
Widely applied criteria, set by different organizations, for
the diagnosis of a syndrome has been detailed out in
supplementary Table 1 (Parikh and Mohan 2012). High fat
high sugar hypercaloric diets (cholesterol and animal fat
plus naturally occurring sugars sucrose and/or fructose)
over time lead to the development of chronic pathologies
like hyperlipidemia, hypercholesterolemia, hyperinsulin-
emia, hyperglycemia, and excess weight causing diabetes,
obesity, hypertension and atherosclerosis in humans and
experimental animal models (Khaled et al. 2012; Nasri
et al. 2018). These diseases are conventionally treated by
various classes of drugs however often adverse and side
effects as well as patient noncompliance and cost issues
impede the treatment.
Evidently, nutrition and natural bioactives may be more
than just food thus offering beneficial health effects upon
intake to the consumer in addition to daily essential nutri-
ents. Once turned down by drug developers, therapeutic
peptides now are ideal candidates for new drug discoveries
(Uhlig et al. 2014). Throughout decades reports have been
compiled about the bioactivities of marine-extracted chem-
icals specifically biopeptides on communicable and
non-communicable diseases. Additionally, in recent years
more attention has been given to anti-MetS phytochemicals,
© 2022 Taylor & Francis Group, LLC
CONTACT Lucie Beaulieu lucie.beaulieu@fsaa.ulaval.ca
Supplemental data for this article is available online at https://doi.org/10.1080/10408398.2022.2052261.
https://doi.org/10.1080/10408398.2022.2052261
KEYWORDS
cardiovascular diseases;
diabetes; sh peptides; metabolic
syndrome; obesity
2 S. ABACHI ETAL.
Table 1. Bioactivities of free amino acids on MetS and associated risk factors.
Amino acid supplement (single or
mixture) Major eect(s) Aected disease (reference)
Diabetes, its risk factors, and associated complications
Aspartate and asparagine insulin sensitivity,
in-vitro glucose uptake by muscle at maximal insulin
concentrations, glucose transport at higher glycogen levels in
rat muscle
(Lancha, Poortmans, and Pereira 2009)
L-alanine and or L-arginine perigonadal and retroperitoneal fat pads,
fed-state glycemia, IRβ, pAS160, glucose tolerance and insulin
secretion in monosodium glutamate-induced obese rats
Diet-induced obesity (Araujo et al. 2017)
BCAA HbA1c of peripheral (primarily muscle) tissue in insulin
resistance chronic hepatitis C patients
Chronic liver diseases (Takeshita etal. 2012)
BCAA glycated albumin and chronic liver disease-HbA1c
post-exercise (enhanced glycemic control) in patients
Liver cirrhosis (Nishida et al. 2017)
BCAA HbA1c and glycoalbumin, fasting plasma glucose, insulin, and
C-peptide (enhanced glycemic control) in nonalcoholic
steatohepatitis-related liver cirrhosis patients
Alternative treatment in absence of eective
conventional therapy for NASH-related
liver cirrhosis (Miyake et al. 2012)
BCAA, arginine, lysine and threonine early postprandial serum insulin response, GLP-1 response,
postprandial glycemia in healthy subjects
(Gunnerud et al. 2012)
Cysteine blood glucose, glycated Hb, CRP, MCP-1, insulin resistance,
plasma protein oxidation, pAkt, and pNF-κB in hypercaloric
fed Zucker diabetic fatty rats
Adjuvant therapy for reduction of vascular
inammation and CVD in diabetics (Jain
et al. 2009) (Jain 2012)
Glutamine FBG, waist circumference,
fat-free mass in T2D patients
(Mansour et al. 2015)
Glutamine dipeptide (Dipeptiven®) hyperglycemic episodes (mean daily insulin dose) in
polytrauma patients
(Grintescu et al. 2015)
Glutamine with or without sitagliptin GLP-1 and insulin secretions,
postprandial glycemia, HbA1c and fructosamine in
well-controlled T2D patients
(Samocha-Bonet et al. 2014) (Greeneld
et al. 2009) (Samocha-Bonet etal. 2011)
Glycine hyperglycemia, cholesterol and glycated hemoglobin, opacity
in lens and microaneurysms in eyes, expression of O-acetyl
sialic acid in brain vessels, intensity of corporal weight loss in
STZ-induced diabetic rat
Diabetic complications: retinopathy, brain
micro-infarcts and renal damage
(Alvarado-Vásquez etal. 2006)
(Alvarado-Vásquez etal. 2003)
Histidine HOMA-IR, BMI, waist circumference, fat mass, serum NEFA and
inammatory cytokines, oxidative stress, GPX,
adiponectin in obese subjects
Metabolic syndrome (Feng et al. 2013)
Hydroxyproline, proline, lysine, glycine,
and alanine
TG accumulation and expression of adiponectin in bovine
retinal pericytes
Protection against proliferative diabetic
retinopathy (Vidhya et al. 2018)
Leucine energy expenditure, fatty acid oxidation and locomotor
activity, expression of UCP-3 in BAT, insulin sensitivity,
intestinal gluconeogenesis,
body weight and fat mass, islets of Langerhans damage,
hepatic lipid in high fat fed rats
Prevention of T2D development (Binder
et al. 2013)
Leucine glucose tolerance, leptin sensitivity during weight
maintenance in obese rats
Adjuvant benecial nutritional therapy
during weight loss and maintenance in
previously diet-induced obesity (Binder
et al. 2014)
Leucine insulin secretion (down-regulation of surface expression of
adrenergic α2A receptor via mTOR) in diabetic Goto-Kakizaki
rats
Clinical management of renal transplant
patients (Yang etal. 2012)
L-leucine, L-lysine, L-isoleucine, L-valine,
L-threonine, L-cysteine, L-histidine,
L-phenylalanine, L-methionine,
L-tyrosine, and L-tryptophan
fasting and postprandial blood glucose and HbA1c, fasting
insulin and insulin resistance,
HDL-c in poorly controlled T2D (HbA1c > 7%) of elderly
subjects
Nutritional supplement for treatment of
elderly T2D diabetes (Solerte et al. 2004)
L-tryptophan and tryptamine in-vitro insulin-stimulated glucose incorporation into
dierentiated adipocyte, in-vivo glucose-associated energy,
serum glucose and insulin, glucose absorption from intestine,
expenditure in T2D rats
Delaying progression of hereditary T2D
(Inubushi et al. 2012)
Lysine blood sugar (maintaining glycosylated hemoglobin and
glycated lens proteins levels), cataract development
Anticataractous in diabetes (Sulochana,
Punitham, and Ramakrishnan 1998)
Methionine blood glucose,
hepatic glycogen in arsenic-induced hypoglycemic rats
Arsenic-induced hypoglycemia and
hypoglycolytic activity management (Pal
and Chatterjee 2004)
Phenylalanine and leucine insulin response following carbohydrate intake in T2D patients Nutritional interventions to improve
postprandial glucose disposal in diabetes
(van Loon et al. 2003)
Serine diabetes incidence and insulitis score, HOMA-IR and blood
glucose, body weight, food, and water intake in non-obese
diabetic rats
Treatment of autoimmune (T1D) diabetes
(Holm et al. 2018)
(Continued)
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 3
Taurine cytosolic [Ca2+]i oscillations in response to stimulatory glucose
concentrations,
insulin secretion at stimulatory glucose concentrations,
glucose metabolism, cytosolic insulin, sulfonylurea receptor-1,
glucokinase, GLUT2, PDX-1 gene expression, nucleus PDX-1
expression, basal and insulin stimulated tyrosine
phosphorylation of insulin receptor in skeletal muscle and
liver tissues, peripheral insulin sensitivity in pancreatic islet
cells of rats
(Carneiro et al. 2009)
Taurine serum glucose and immunoreactive insulin,
glucose clearance, deoxyglucose accumulation in skeletal
muscle and liver, hepatic glycogen synthesis in rats
(Ito, Schaer, and Azuma 2012; Kulakowski
and Maturo 1984)
Taurine hyperglycemia in alloxan-induced T1D rabbits (Tenner, Zhang, and Lombardini 2003)
Taurine lectin-type oxidized LDL receptor 1 and ICAM-1 expression on
aortas (improving vascular endothelial dysfunction) in
streptozotocin-induced T1D rats
(L.-j. Wang et al. 2008)
Hypercholesterolemia, its risk factors, and associated complications
Arginine with simvastatin serum TG in hypertriglyceridemic patients Lipid metabolism (Schulze et al. 2009)
Arginine, alanine, glycine serum cholesterol and extent of aortic sudanophilia in
casein-induced hypercholesterolemic and atherosclerotic
rabbits and rats
(Katan, Vroomen, and Hermus 1982)
L-aspartate and l-glutamate serum HDL-c, maintaining ApoA-1 in hypercholesterolemic
diet fed rabbits
Antiatherogenic (Yanni, Perrea, and Yatzidis
2005)
BCAA HOMA-IR, insulin, hsCRP, waist/height ratio and SBP in twin
subjects
Eects on cardiometabolic health (Jennings
et al. 2016)
Histidine food intake, retroperitoneal fat pad weight, fat accumulation,
brown adipose tissue UCP-1 mRNA in rats
(Kasaoka et al. 2004)
Histidine, isoleucine, leucine, lysine,
methionine, phenylalanine,
threonine, valine, and arginine
plasma TG, TC, and VLDL-c, hepatic fat in impaired glucose
tolerant elderly patients
Treatment of hypertriglyceridemia or hepatic
steatosis (Børsheim et al. 2009)
(Abete et al. 2010)
Isoleucine body weight gain, eWAT mass, hepatic and skeletal muscle
TG, hyperinsulinemia, WAT leptin,
WAT adiponectin, hepatic levels of protein CD36/fatty acid
translocase, PPARα, and UCP-2, skeletal muscle levels of
UCP-3 in high fat fed obese rats
(Nishimura et al. 2010)
Leucine appendicular muscle mass during intentional weight loss in
obese older adults
Reducing sarcopenia risk in obesity
(Verreijen et al. 2015)
Leucine hepatic steatosis, adipose tissue inammation, subcutaneous
fat pad and liver weight, insulin resistance, glucose
intolerance, expression of lipogenic genes, hepatic lipid
deposition,
insulin-stimulated phosphorylation of p70S6 kinase, activation
of mTOR in high fat fed obese rats
Adjunct therapy in management of
obesity-related insulin resistance
(Macotela et al. 2011)
Methionine plasma homocysteine, cholesterol in plasma and liver, hepatic
expression of HMG-CoA reductase and CYP7A1,
phosphatidylcholine: phosphatidylethanolamine ratio in liver
of rats
Methionine-induced hypercholesterolemia
via enhanced hepatic cholesterol
synthesis (Hirche et al. 2006)
Phenylalanine, arginine, and alanine serum glycerol, FFA and acetoacetic acid in healthy subjects Stimulating fat metabolism during exercise
(Ueda et al. 2016)
Taurine liver weight and liver weight/body weight ratio, plasma TC,
glucose and LDL-c, hepatic cholesterol, and TG,
HDL-c in hypercholesterolemic diet fed rats
Hypercholesterolemia (M.-J. Choi, Kim, and
Chang 2006)
Taurine and n-3 FA TC, LDL-c, ApoB, TG, thromboxane B (2), TNF-α, and MCP-1,
HDL-c in healthy subjects
Hypolipidemic and antiatherogenic eects
of seafood diet (Elvevoll et al. 2008)
Threonine hepatic TL, TG, TC, and lipid deposition in Pekin ducks (Jiang et al. 2017)
Valine and leucine serum SOD and GPx in hypercholesterolemic rats Improving atherosclerosis-associated
endothelial dysfunction (Cojocaru et al.
2014)
Weight management, obesity, and its related complications
Alanine and or leucine body fat accumulation, hepatic TG, expression of lipogenic
enzymes and plasma cholesterol in high fat diet induced
obese rats
Metabolic syndrome (Freudenberg, Petzke,
and Klaus 2013; K. Petzke, Freudenberg,
and Klaus 2014)
Arginine serum TG, fat mass,
serum glucagon, body weight gain and skeletal-muscle mass
in growing-nishing pigs
Obesity therapeutic development (Tan etal.
2009)
Arginine weight of abdominal and epididymal adipose tissues, serum
glucose, TG, FFA,
lipolysis and glucose oxidation in abdominal and epididymal
adipose tissues of diabetic obese Zucker rats
Obesity (Fu et al. 2005)
BCAA BMI and waist circumference, prevalence of overweight/
obesity, abdominal obesity, postprandial glucose tolerance
and status of inammation in young subjects
Prevention of obesity (Li et al. 2015)
BCAA food intake and weight gain,
insulin resistance in high fat fed rats
Obesity-associated BCAA-induced insulin
resistance (Newgard et al. 2009)
Table 1. (Continued).
Amino acid supplement (single or
mixture) Major eect(s) Aected disease (reference)
(Continued)
4 S. ABACHI ETAL.
BCAA body weight and eWAT mass, hepatic, and skeletal muscle TG,
hepatic expression of PPARα and UCP-2, skeletal muscle
expression of PPARα and UCP-3 in high fat fed obese rats
(Arakawa et al. 2011)
BCAA prevalence of overweight/obesity in healthy East Asian and
Western middle-aged subjects
(Qin et al. 2011)
EAA lean body mass, basal muscle protein synthesis, and IGF-1
expression in elderly subjects
(Dillon et al. 2009)
Glutamine body weight, waist circumference, insulinemia and HOMA-IR
in obese subjects
Enhancing glucose metabolism and weight
loss (Laviano et al. 2014)
Glutamine body weight, plasma glucose and insulin in high fat fed
hyperglycemic and hyperinsulinemic overweight rats
Obesity and diabetes amelioration in
pre-diabetic and diabetic state (Opara
et al. 1996)
Glycine weight loss and mRNA expression of metabolic involved
genes,
whole-body and epididymal fat mass, glucose intolerance,
mRNA expression of pro-inammatory involved genes, S6
protein phosphorylation in high fat induced obese rats
Accelerating loss of adipose tissue and
protecting muscle mass in obesity
treatment (Caldow etal. 2016)
Histidine energy intake,
FFA, lipolysis in moderately obese subjects
Weight loss in moderate obesity (Konomi
et al. 2004)
L-arginine maternal lipid and adiposity and circulating leptin,
fetal brown adipose tissue development in diet-induced obese
sheep
Maternal obesity (Sattereld et al. 2012)
L-arginine adiposity, WAT, and fat mass,
mitochondrial biogenesis and brown adipose tissue
development, muscle mass, expression of glucose and FA
oxidation involved genes in obese rats, nishing pigs and
T2D patients
Reducing obesity (McKnight et al. 2010)
Leucine weight gain and adiposity, hyperglycemia, plasma glucagon
and glucogenic AA, hepatic glucose-6-phosphatase, plasma
TC and LDL-c
resting energy expenditure, expression of UCP-3 in brown and
white adipose tissues and in skeletal muscle, insulin
sensitivity in high fat fed obese rats
(Yao et al. 2016; Y. Zhang et al. 2007)
Leucine peripheral fat oxidation and glucose transport (independent
of appetite and weight regulation) in ospring from high fat
fed obese mother rats
Management of metabolic disorders and
maternal obesity and (Chen et al. 2012)
Leucine glycemic control, insulin response to food challenge and
insulin, energy expenditure, skeletal muscle expression of
genes involved in regulating energy metabolism (UCP-3,
carnitine acetyltransferase, PPARα, and NRF-1),
HbA1c, adipose tissue inammation in obese and T2D rats
Dietary intervention in prevention and
management of obesity and T2D (Guo
et al. 2010)
L-phenylalanine insulin, GLP-1, and peptide tyrosine tyrosine (PYY) release
food intake (via blockade of calcium-sensing receptor), plasma
ghrelin, glucose intolerance in rats
Treatment of obesity and diabetes
(Alamshah et al. 2017)
Lysine, proline, alanine, and arginine
with or without conjugated linoleic
acid
body weight and fat, BMI, waist, and hip circumferences
after a period of daily exercise in healthy overweight subjects
Reducing visceral fat and enhancing
fat-burn during exercise, prevention of
metabolic syndrome (Michishita et al.
2010)
Taurine glucose intolerance, glucagon-induced hepatic glucose output,
insulin hypersecretion and hyperinsulinemia, pancreatic islet
hypertrophy,
glucose homeostasis, pancreatic islet-cell morphology, and
function in leptin-decient genetically obese rats
Management of insulin hypersecretion and
hyperinsulinemia in obesity and T2D
(Santos-Silva et al. 2015)
Tryptophan weight loss in moderately obese patients (Heraief et al. 1985)
Tryptophan and lysine food intake and body weight in rats (Ayaso etal. 2014)
Tyrosine food intake of nocturnal period, fasting plasma insulin,
food intake of diurnal period, fasting plasma glucose in rats
(Bassil, Hwalla, and Obeid 2007)
IRβ: Insulin receptor β, pAS160: Phosphorylated Akt substrate of 160 kDa, pAkt: Phosphorylated Akt, pNF-κB: Phosphorylated NF-κB, PDX-1: Proconvertase and
pancreas duodenum homeobox-1, BMI: Body mass index, HDL-c: High-density lipoprotein-cholesterol, TG: Triglyceride.
food-derived lipids, fibers as well as peptides including
amino acids and their regulatory effects on the syndrome’s
main risk factors (Table 1) (Cicero and Colletti 2016;
Delzenne and Cani 2005; Iwaniak, Darewicz, and Minkiewicz
2018; Nagao and Yanagita 2008; Power et al. 2014;
Ricci-Cabello, Olalla Herrera, and Artacho 2012; Xia etal.
2017). As discussed in this review, anti-obesity, anti-diabetes
and anti-MetS fish biopeptides could be extracted by dif-
ferent methods and techniques. Peptides with anti-MetS
attributes have been successfully extracted from fish, fish
products and byproducts by mainly enzymatic hydrolysis.
Nevertheless, atypical extraction methods such as subcritical
water extraction, isoelectric precipitation and or microwave
pretreatment have as well been practiced. All sorts of fil-
tration procedures, micro-, nano- and ultra-filtration, are
typically exercised before chromatographic peptide purifi-
cation techniques (Table 2). For characterization purposes
of fish anti-MetS peptides, chromatography, spectrophotom-
etry, and electrophoresis techniques are frequently in use
(Table 2).
Table 1. (Continued).
Amino acid supplement (single or
mixture) Major eect(s) Aected disease (reference)
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 5
Table 2. Techniques used for extraction, isolation, purication, and characterization of anti-MetS biopeptides from sh and sh-derived products.
Fish name (common,
scientic) and part used
Hydrolysis, fractionation,
and purication Condition and resin/material Characterization References
Alaska pollack (Theragra
chalcogramma) llet
1- enzymatic hydrolysis,
2- RP-HPLC
1- trypsin,
2- octadecyl silica (ODS) Cosmosil
5 C18 column
MS, protein sequencer, UPLC
(Acquity BEH-C18 RP
column) coupled to Xevo
Q-TOF–MS equipped with
electrospray ionization
(ESI) source
(Ayabe etal. 2015)
Alaska pollack (Theragra
chalcogramma) llet
1- enzymatic hydrolysis 1- pepsin (Mizushige et al.
2017)
Alaska pollock (Theragra
chalcogramma) llet
1- dehydration 1- N2 gas (Hosomi et al.
2013) (Hosomi
et al. 2011)
Alaska pollock (Theragra
chalcogramma) llets
1- our preparation 1- lyophilization HPLC, SDS-PAGE (Maeda etal. 2015)
Allaska pollock (Theragra
chalcogramma)) llet
1- enzymatic hydrolysis 1- papain SDS-PAGE, HPLC, gel
permeation HPLC (TSK
gel column)
(Hosomi et al.
2010; Hosomi
et al. 2012)
Atlantic cod (Gadus
morhua),
commercial preparation by
Firmenich Bjørge
Biomarin AS, Norway
1- enzymatic hydrolysis 1- protamex (Dale et al. 2018)
Atlantic herring (Clupea
harengus) byproducts
(heads, guts, and
backbones), Atlantic
salmon (Salmo salar)
byproducts (backbones)
1- enzymatic hydrolysis 1- papain and bromelain (Drotningsvik,
Pampanin, et al.
2018)
Atlantic herring (Clupea
harengus) head, gut,
backbone byproducts,
Atlantic salmon, (Salmo
salar) backbone
byproduct, Atlantic cod
(Gadus morhua) muscle,
commercial tablet
preparation by Faun
Pharma AS, Vestby,
Norway
1- enzymatic hydrolysis,
2- cod our preparation
(cooking, drying and
micro-milling),
3- dosage preparation
1- herring and salmon (mixture of
papain and bromelain),
2- −
3- tableting
HPLC (Hovland etal.
2019)
Atlantic salmon (Salmo
salar) fresh skin
byproducts (recovered
from skin-o llets)
1- aqueous gelatin
extraction,
2- enzymatic hydrolysis,
3- UF,
4- HPLC
1- distilled, deionized water (ddH2O),
2- alcalase (DH: 35 41%), bromelain
(DH: 28 38%), avourzyme (DH:
42%),
3- MWCO 2.5 − 1 kDa,
4- Zorbax Eclipse Plus C18 column
Amino acid analyzer,
Q-TOF-MS-ESI
(Li-Chan et al.
2012)
Atlantic salmon (Salmo
salar) fresh skin
byproducts (recovered
from skin-o llets)
1- aqueous gelatin
extraction,
2- enzymatic hydrolysis
1- distilled, deionized water (ddH2O),
2- avourzyme
(Hsieh et al. 2015)
Atlantic salmon (Salmo
salar) frozen frames
1- alkaline solubilization,
2- enzymatic hydrolysis,
3- UF,
4- RP-HPLC
1- NaOH
2- pepsin, tr ypsin + chymotrypsin,
3- MWCO 1 kDa,
4- C12 column
(Chevrier et al.
2015)
Atlantic salmon (Salmo
salar) skin and
trimmings byproduct
1- aqueous gelation
extraction,
2- enzymatic hydrolysis
1- 1:5 H2O (w/v) (skin only)
2- alcalase (DH: skin and trimming,
DH: 11% and 27%,
alcalase + avourzyme (DH: 17%
and 34%), promod (DH: 6% and
27%)
Gel permeation (GP)-HPLC,
RP-UPLC, UPLC-ESI-MS/
MS coupled to impact
HD ultra-high resolution
(UHR) Q-TOF
(Harnedy et al.
2018a)
Barbel (Barbus callensis)
fresh muscle
1- enzymatic hydrolysis,
2- SEC, RP-HPLC, HPLC
1- alcalase (DH: 16.4%),
2- gel ltration Superdex peptide
10/30 column, TRACER-Excel 1200
DS-A column, HALO Peptide
ES-C18 column
ESI-MS, and ESI-MS/MS (Assaad Sila et al.
2016)
Barbel (Barbus callensis)
fresh skin
1- enzymatic hydrolysis 1- barbel crude acid protease
(limited gelatin hydrolysis),
esperase (DH: 9.3%), savinase (DH:
9.2%), alcalase (DH: 14.2%),
trypsin (DH: 8.5%), izyme G (DH:
5.8%), protamex (DH: 10.4%),
neutrase (DH: 9.4%), peptidase
(DH: 7.4%)
Amino acid analyzer,
SDS–PAGE
(Assaâd Sila et al.
2015)
(Continued)
6 S. ABACHI ETAL.
Bester sturgeons (Huso
huso × Acipenser
ruthenus) skin, n, and
bone
1- collagen extraction,
2- collagen enzymatic
hydrolysis,
3- atelocollagen extraction
4- atelocollagen enzymatic
hydrolysis,
5- gel ltration
chromatography, RP-HPLC
1- salt (1% NaCl), acid (0.2 M HCl),
alkali (0.2 M NaOH),
2- papain,
3- 5% H2O2, salt (1% NaCl), acid
(0.2 M HCl), alkali (0.2 M NaOH),
99.5% ethanol,
4- pepsin,
5- Sephadex G50 and Sephadex
G25, Mightysil RP18 GP Aqua
column
RP-HPLC (protein sequencer
Procise 493), SDSPAGE
(Sasaoka et al.
2018)
Bighead carp
(Hypophthalmichthys
nobilis) muscle
1- enzymatic hydrolysis,
2- UF,
3- gel ltration
chromatography,
semi-preparative RP-HPLC
1- papain (DH: 6 8%), alcalase (DH:
6 − 10%), tr ypsin (DH: 11 − 13%),
pepsin (DH: 14 − 16%),
2- MWCO 5 − 3 kDa,
3- Sephadex G-15, Kromasil 100-5-C18
semi-preparation column
Size exclusion (SE)-HPLC
(TSK gel G2000 SWXL
column), LC-MS/MS (RP
capillary column)
(C. Zhang et al.
2017)
Blacktip shark (Carcharhinus
limbatus) skin byproduct
1- aqueous gelatin
extraction,
2- enzymatic hydrolysis
1- H2O,
2- crude enzyme from papaya
(Carica papaya) latex (DH: 10, 20,
30 and 40%)
(Kittiphattanabawon
et al. 2013)
Blue whiting
(M. poutassou), fresh frozen
muscle, commercial food
supplement preparation
containing Slimpro® by
Compagnie des Pêches
Saint Malo Santé, France
1- enzymatic hydrolysis 1- − (Nobile et al. 2016;
Zaïr et al. 2014)
Blue whiting
(Micromesistius
poutassou) fresh frozen
muscle
1- enzymatic hydrolysis
(pH-stat method)
1- alcalase (DH: 18.8%) (Cudennec et al.
2012)
Blue whiting
(Micromesistius
poutassou) muscle
1- enzymatic hydrolysis 1- alcalase + avourzyme (DH: 29%) HPLC, amino acid analyzer,
UPLC-ESI-MS/MS coupled
to impact HD UHR Q-TOF
(Harnedy et al.
2018b)
Blue whiting fresh frozen 1- aqueous soluble protein
extraction (cooking,
dewatering, sieving,
centrifugation and
drying)
1- H2O(Drotningsvik,
Vikøren, et al.
2018)
Boarsh (Capros aper)
muscle
1- enzymatic hydrolysis 1- alcalase + avourz yme RPUPLC (Acquity BEH
300 C18 RP column),
GPHPLC (TSK G2000 SW
separating and guard
columns)
(Parthsarathy et al.
2019)
Bogue (Boops boops) llet 1- enzymatic hydrolysis 1- crude alkaline proteases from
smooth hound viscera (DH: 20%)
HPLC (AccQ.Tag amino acid
analyzing column
(Nova-Pak C18 column)
(Lassoued et al.
2014)
Cod (Theragra
chalcogramma) and
tuna (Thunnus orientalis)
light muscle
1- dehydration 1- − HPLC, SDS-PAGE (Hosomi etal.
2017)
Cod llet 1- freeze drying and milling 1- − (Myrmel et al.
2019)
Crucian carp (Carassius
carassius) muscle (head
and viscera excluded)
1- enzymatic hydrolysis 1- neutral protease (DH: 20%),
alkaline protease (DH 25%),
papain (DH: 20%), and protamex
(DH: 20%)
(L. Liu et al. 2013)
Fish (lean-sh cod, pollock
and haddock skin),
commercial preparation
of high MW food/
pharmaceutical-grade sh
gelatin by Norland
Products Incorporated,
United States
1- enzymatic hydrolysis
(collagen extraction)
1- − (Picard-Deland
et al. 2012)
Fish scales, commercial
collagen preparation by
Nippi Co. Ltd., Japan
1- enzymatic hydrolysis 1- Streptomyces collagenase, DPP-IV
from Pichia pastoris
Gel ltration
chromatography
(Superdex peptide
column), HPLC
(Hypercarb column)
coupled with evaporative
light scattering detector
(ELSD)
(Hatanaka,
Kawakami, and
Uraji 2014)
Table 2. (Continued).
Fish name (common,
scientic) and part used
Hydrolysis, fractionation,
and purication Condition and resin/material Characterization References
(Continued)
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 7
Fresh Norwegian Atlantic
salmon o-cut
byproducts (spine),
commercial 91% pure
protein preparation by
Marine Bioproducts AS,
Norway
1- enzymatic hydrolysis,
2- ltration (micro- and
ultra-)
1- alkaline and neutral proteases
2- −
(Bjørndal et al.
2013; Parolini
et al. 2014)
Goby sh (Zosterisessor
ophiocephalus) fresh
muscle
1- enzymatic hydrolysis 1- Bacillus mojavensis A21 (DH:
13.4%), gray triggersh digestive
crude proteases (DH: 23.4%)
HPLC (AccQ·Tag amino acid
analyzing Nova-Pak C18
column)
(Nasri et al. 2018;
Nasri et al.
2015)
Gray triggersh (Balistes
capriscus) muscle
1- enzymatic hydrolysis 1- crude enzyme preparations from
Bacillus mojavensis A21,
sardinelle (Sardinella aurita)
viscera, Zebra blenny (Slaria
basilisca) viscera
(Siala et al. 2016)
Herring milt hydrolysate
commercial preparation
1- enzymatic hydrolysis,
2- UF,
3- electro-separation
1- −
2- −
3- EDUF 50, 20 kDa
(Durand et al.
2019)
Mixture of wild caught cod
llet and Canadian
scallop muscles (cod/
scallop, 1 : 1 on amino
acid content)
1- freeze drying and milling 1- − (I.-J. Jensen et al.
2016; Tastesen
et al. 2014)
Nile tilapia (Oreochromis
niloticus) fresh
skin
1- alkaline and aqueous
collagen extraction,
2- enzymatic hydrolysis
1- 0.05 M NaOH, H2O, 1% Ca(OH)2,
2- alcalase
Automatic amino acid
analyzer, HPLC (TSK gel
column)
(R. Zhang, Chen,
Chen, et al.
2016)
Norwegian spring spawning
herring and salmon
(backbones) fresh
byproducts
1- enzymatic hydrolysis 1- papain and bromelain (Drotningsvik et al.
2016)
Pacic hake (Merluccius
productus), halibut
(Hippoglossus
stenolepis), tilapia
(Oreochromis niloticus),
milksh (Chanos chanos)
fresh byproduct skins
recovered from skin-o
llets
1- aqueous gelatin
extraction,
2- enzymatic hydrolysis,
3- UF
1- −
2- avourzyme,
3- MWCO 2.5 − 1.5 kDa
(T.-Y. Wang et al.
2015)
Pollock (Theragra
chalcogramma) esh
and frame byproduct
1- enzymatic hydrolysis,
2- UF
1- alcalase and avourzyme,
2- MWCO 1 kDa
SEC (TSK G2000 column) (Cai et al. 2015; Xu
et al. 2016;
Zheng et al.
2014)
Rainbow trout
(Oncorhynchus mykiss)
fresh frames
1- microwave assisted
extraction,
2- enzymatic hydrolysis,
3- electrodialysis with
ultraltration membrane
(EDUF)
1- microwave pretreatment (90 °C,
10 min, 800 W ),
2- alcalase,
3- anionic and cationic
exchange membranes, MWCO
20 kDa
MALDIMS (Ketnawa et al.
2019)
Salmon (Salmo salar) and
cod (Gadus morhua)
frozen byproduct frames
1- alkaline solubilization,
2- aqueous dispersion,
3- enzymatic hydrolysis,
4- UF
1- 0.1 and 1 M NaOH,
2- dH2O,
3- pepsin, pancreatin, trypsin and
chymotrypsin (alone or in
combination), 4- MWCO 1 kDa
(Jin 2013)
Salmon (Salmo salar)
trimmings
1- enzymatic hydrolysis,
2- semi-preparative RP-HPLC
1- dierent preparations and 1-,
2- and 4-h durations (alcalase
(DH: 14.0 − 18.3%),
alcalase + avourzyme (DH:
15.1 − 16.0%), corolase (DH:
21.8 − 24.3%), PROMOD (DH:
20.3 − 22.1%),
2- C18 semi-preparative column
SDS-PAGE, RP-HPLC, gel
permeation HPLC,
UPLC-MS/MS (Peptide
XB-C18 column)
(Neves et al. 2017)
Salmon frame byproducts 1- alkaline solubilization,
2- isoelectric precipitation,
3- enzymatic hydrolysis,
4- UF,
5- EDUF,
1- NaOH (1 N),
2- pH of 4.5,
3- pepsin, trypsin/chymotrypsin,
4- MWCO 1 kDa,
5- MWCO 50, 20, 5 kDa
RP-UPLC-MS/MS (C18
column)
(Henaux et al.
2019)
Table 2. (Continued).
Fish name (common,
scientic) and part used
Hydrolysis, fractionation,
and purication Condition and resin/material Characterization References
(Continued)
8 S. ABACHI ETAL.
Salmon frames 1- alkaline solubilization,
2- isoelectric precipitation,
3- enzymatic hydrolysis,
4- UF, EDFM
1- 1 M NaOH,
2- pepsin at pH 4.5,
3- trypsin and chymotrypsin,
4- prep/scale tangential ow
ltration (TFF) cartridge MWCO
1 kDa, Neosepta CMX-SB cationic
and AMX-SB anionic membranes,
UF polyether sulfone (PES) MWCO
of 20 kDa
RP-HPLC and HPLC/MS (BEH
C18 column), HPLC
(AccQTag amino acid
analysis column (silica
base bonded with C18))
(Roblet et al. 2016)
Salmon fresh backbones 1- enzymatic hydrolysis 1- corolase PP (DH: 18 22%) and
corolase 7089 (DH: 14 18%),
protamex (DH: 15 − 21%),
papain + bromelain (DH: 13 − 17%),
trypsin (DH: 14 18%), protex 6 L
(DH: 14 18%), seabzyme L200
(DH: 13 − 17%)
Gel ltration on FPLC
(Superdex™ Peptide
10/300 GL column), HPLC
(Slizyte et al. 2016)
Salmon spine and or
backbones including
heads, commercial
preparation by Marine
Bioproducts AS, Norway
1- enzymatic hydrolysis,
2- ltration (micro- and
ultra-)
1- salmon spine: alkaline and neutral
protease with sequential
hydrolysis (acid protease A and or
umamizyme of Aspergillus
oryzae), salmon backbones
including heads: alcalase
2- −
(Vik et al. 2015)
Sardine (S. pilchardus) and
sardinelle (S. aurita)
fresh muscle
1- enzymatic hydrolysis 1- alcalase (DH: 8%) (Athmani et al.
2015)
Sardine (Sardina pilchardus)
and bogue (Boops
boops) fresh muscle
1- enzymatic hydrolysis 1- alcalase (DH: 8%) HPLC (Benomar et al.
2015)
Sardine (Sardina pilchardus)
fresh byproducts (viscera,
heads, skins and edges)
and llets
1- our preparation,
2- isoelectric precipitation
1- cooking, oven drying and hexane
dilapidation
2- 98% H2SO4
(Aane et al. 2018)
Sardine (Sardina
pilchardus), horse
mackerel (Trachurus
mediterraneus), axillary
seabream (Pagellus
acarne), bogue (Boops
boops), small-spotted
catshark (Scyliorhinus
canicula), blue whiting
(Micromesistius
poutassou) west
Mediterranean Sea sh
discards
1- enzymatic hydrolysis
(sequential 2-stage and
1-stage)
1- sequential 2-stage (1st: subtilisin,
2nd: trypsin (DH: 14.9 19.7%) and
(1st: trypsin, 2nd: subtilisin (DH:
13.2 − 18.3%)), 1-stage
(combination of subtilisin + tr ypsin
(DH: 13.7 − 21.0%))
(Pérez-Gálvez et al.
2015)
Sardine llet
muscle tissue
1- alkaline solubilization 1- NaOH (Madani et al.
2015)
Sardine llet (head, internal
organs and bones
excluded)
1- acid and alkaline
solubilization
1- 2 N HCl, 2 N NaOH (Madani et al.
2012)
Sardinelle (Sardinella
aurita) fresh muscle
1- meat our preparation,
2- fermentation
1- cooked and oven dehydrated,
2- Bacillus subtilis A26 (DH: 21%)
and B. amyloliquefaciens An6
(DH: 24%)
RP-HPLC (Symmetry C18
column, Pico Tag
column), MALDI-TOF-MS
(Jemil, Abdelhedi,
et al. 2017;
Jemil, Nasri,
et al. 2017)
Sardinelle (Sardinella
aurita) fresh muscle
1- enzymatic hydrolysis Bacillus pumilus A1 (DH: 14%),
Bacillus mojavensis A21 (DH:
7.5%), sardinelle viscera (DH:
8.5%) crude proteases extract
(Khaled et al. 2012)
Shark (Squalus mitsukurii
and Chiloscyllium
plagiosum) fresh healthy
liver
1- cooking,
2- UF,
3- IEC, gel chromatography,
FPLC, RP-HPLC
1- −
2- MWCO 30 kDa,
3- DEAESepharose column, BioGel
P10 column, monoQ column, C18
column
SDSPAGE, MALDIMS,
automatic amino acid
analyzer
(F. J. Huang & Wu,
2010)
Siki (Centroscymnus
coelolepis) cooked head
and saithe (Pollachius
virens) cooked muscle
1- enzymatic hydrolysis,
2- UF,
3- deodorization,
4- SEC, RP-HPLC
1- siki: alcalase, saithe: commercial
proteases,
2- MWCO 10 kDa,
3- active carbon,
4- HW-40 Toyopearl column,
Prosphere 300 C18 column
Protein sequencer, SEC
(Superdex peptide PC
3.2/30 column),
LC-ESI-IT-MS/MS (RP trap
column (PepMap C18
-precolumn) and ACE
3 m C18 RP analytical
column)
(Martínez-Alvarez
et al. 2012)
Table 2. (Continued).
Fish name (common,
scientic) and part used
Hydrolysis, fractionation,
and purication Condition and resin/material Characterization References
(Continued)
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 9
Silver Carp
(Hypophthalmichthys
molitrix Val.) fresh
muscle
1- enzymatic hydrolysis,
2- UF,
3- thin layer
chromatography, RP-HPLC
1- alcalase (DH: 29%), papain (DH:
25%), neutrase 20%),
avourzyme, trypsin and pepsin
(DH: 11 − 12%),
2- MWCO 10, 5, and 3 kDa,
3- ready-use silica gel 60 plate,
analytical C18 column
LC-ESI-MS/MS (Ying Zhang, Chen,
Chen, et al.
2016)
Skate (Raja Kenojei) skin,
collagenous preparation
to capsule form by
Serom Co., Ltd.
(Jeonnam, South Korea)
1- enzymatic hydrolysis,
2- dosage preparation
1- −
2- capsuling
(Tak et al. 2019)
Skate skin 1- collagen extraction 1- − (H. Lee et al. 2018)
Skipjack Tuna (Katsuwonus
Pelamis) heart
1- ethanolic extraction 1- 70% ethanol (Ali et al. 2016)
Smooth hound (M.
mustelus) muscle
1- enzymatic hydrolysis 1- crude smooth hound intestinal
proteases (DH: 20.3%)
ESI-MS, GC/MS (CPSil5 CB
Low bleed/MS capillary
column)
(Bougatef et al.
2010)
Smooth hound viscera 1- enzymatic hydrolysis,
2- successive UF
1- Purafect® (DH: 14.5%),
2- MWCO 1 kDa
RP-HPLC (PicoTag® column) (Abdelhedi et al.
2019)
Steelhead (Oncorhynchus
mykiss) fresh skin gelatin
1- enzymatic hydrolysis,
2- UF
1- alcalase, bromelain, papain,
protease P “Amano” 6SD, protease
M “Amano” SD, sequentially
hydrolyzed with corolaseN and or
pepsin,
2- MWCO 3 kDa
(Cheung and
Li-Chan 2017)
Thornback ray (Raja
clavata) fresh muscle
1- enzymatic hydrolysis 1- mixture of neutrase, alcalase,
crude proteases of B. subtilis A26
and R. clavata
(Lassoued et al.
2018)
Tilapia (Oreochromis sp.)
scale, commercial food
grade collagen (Wellnex®
Type D) preparation by
Nitta Gelatin, Inc., Japan
1- enzymatic hydrolysis (Iba et al. 2016)
Tuna (Thunnus tonggol)
cooking juice (5.44%
protein)
1- enzymatic hydrolysis,
2- gel ltration
chromatography, RP-HPLC
1- protease XXIII (PR) (DH: 19.4%)
and orientase (OR) (DH: 23.6%),
2- Sephadex G-25 column, Zorbax
Eclipse Plus C18 column
MALDI-TOF/TOF MS/MS (S.-L. Huang et al.
2012)
Tuna extract 1- desalination and boiling,
2- ltration,
3- precipitation (soluble and
insoluble proteins),
4- HPLC
1- −
2- MWCO 200 Da,
3- methanol, chloroform, and water,
4- C18 column
SDS-PAGE, Q-TOF MS/MS (Y. M. Kim etal.
2015)
Tuna extract 1- desalination and boiling,
2- ltration,
3- sterilization
1- −
2- MWCO 200 Da
3- −
(Y. Kim etal. 2016)
Tuna skin byproduct 1- subcritical water
extraction
1- 190 °C and pressure chamber of
1100 kPa
(E. J. Lee et al.
2017)
Unicorn leatherjacket sh
(Aluterus monoceros)
skin byproduct
1- enzymatic hydrolysis,
2- UF, tangential ow
ltration
1- crude collagenase enzyme from
sh ns (DH: 7.6%) (at 5, 25,
50 °C temperatures),
2- MWCO 30 kDa, MWCO 10, 3 kDa
(Kumar, Shakila,
and Jeyasekaran
2019)
Warm sea sh skin,
commercial type I and III
collagen preparation
(Naticol®) by Weishardt,
France
1- enzymatic hydrolysis 1- − (Astre et al. 2018)
Wild marine deep-sea sh
fresh meat, commercial
preparation
1- enzymatic hydrolysis 1- mixed proteases (C.-F. Zhu, Li, Peng,
Li, et al. 2010)
Wild marine sh 1- enzymatic hydrolysis
(collagen extraction)
1- mixed proteases (25% pepsin +
35% trypsin + 35% chymotrypsin
+ 5% pancreatic lipase)
HPLC, MALDI-TOF-MS,
H835-50 automatic
amino acid analyzer
(C.-F. Zhu, Li, Peng,
Zhang, et al.
2010)
Wild marine fresh sh 1- enzymatic hydrolysis,
2- UF,
3- desalination,
4- cryoconcentration,
5- decolorization and
lyophilization
1- mixed proteases,
2- −
3- −
4- 70 °C under vacuum,
5- −
HPLC, MALDI-TOF-MS,
automatic amino acid
analyzer
(Cui-Feng etal.
2010)
Table 2. (Continued).
Fish name (common,
scientic) and part used
Hydrolysis, fractionation,
and purication Condition and resin/material Characterization References
(Continued)
10 S. ABACHI ETAL.
Wild marine fresh sh scales 1- demineralization,
2- collagen extraction,
3- salting out precipitation,
4- dialysis,
5- enzymatic hydrolysis,
6- ltration
1- 0.4 M HClO4,
2- 0.5 M acetic acid (+ 0.005 M EDTA),
3- 0.9 M NaCl concentration,
4- against distilled water,
5- pepsin,
6- ceramic membrane (200 m
26 kDa)
(Raksha et al. 2018)
Wild-caught chum salmon
skin
1- collagen extraction,
2- enzymatic hydrolysis,
3- UF,
4- HPLC,
5- desalination,
6- decolorization
1- −
2- mixed proteases,
3- MWCO 10 kDa,
4- C18 column,
5- −
6- medicinal charcoal
MALDI-TOF-MS, automatic
amino acid analyzer,
HPLC
(C. Zhu, Zhang, Mu,
et al. 2017)
Wild-caught chum salmon
skin
1- collagen extraction,
2- enzymatic hydrolysis,
3- spray drying
1- −
2- alcalase, protamex
3- −
HPLC, MALDI-TOF-MS,
automatic amino acid
analyzer
(C. Zhu, Zhang, Liu,
et al. 2017)
Wild-caught chum salmon
skin
1- collagen extraction 1- − (J. Wang et al.
2010)
Yellown tuna (Thunnus
albacares) fresh bone
1- alkaline, acid, and
aqueous solubilization
(collagen extraction),
2- enzymatic hydrolysis
1- 0.1 M NaOH, 1.5% acetic acid and
aquadest,
2- collagenase from Bacillus sp. 6-2
(Natsir, Dali, and
Arif 2019)
Zebra blenny (Salaria
basilisca) fresh and or
fresh frozen muscle
1- enzymatic hydrolysis 1- crude proteases from viscera of
zebra blenny (DH: 10%), smooth
hound (Mustelus mustelus) (DH:
9.1%), sardinelle (S. aurita) (DH:
6.65%)
HPLC (AccQ·Tag amino acid
analyzing Nova-Pak C18
column), SEC (Sephadex
G-25 column)
(Ktari et al. 2015;
Ktari et al. 2013)
Zebra blenny (Salaria
basilisca) fresh muscle
1- enzymatic hydrolysis 1- crude alkaline proteases from
homogenized viscera of zebra
blenny, sardinella and smooth
hound
HPLC-SEC (PL aquagel-OH
MIXED-H
preparative-column)
(Ktari et al. 2017)
Table 2. (Continued).
Fish name (common,
scientic) and part used
Hydrolysis, fractionation,
and purication Condition and resin/material Characterization References
Amino acids, mixture or single, or peptides as mono-
and or combination therapies with clinical drugs, have
shown promising health effects on MetS throughout many
studies in animals, humans and in cell analysis (Table 1).
Dietary amino acid supplementation had glucoregulatory
and hypocholesterolemic effects among human subjects with
spinal cord injury (glucoregulatory activity of all amino
acids, in particular lysine, exclusive of cysteine, glutamic
acid, threonine, leucine and histidine on fasting plasma
glucose as well as hypocholesterolemic activity of threonine
and leucine on TG and total cholesterol (TC)) (Javidan
et al. 2017). Fermented mackerel extract and its fractions,
free amino acid deficient NH4OH-HCl fraction (3.2%) and
free amino acid rich acetonitrile (ACN) fraction (94.5%),
with various peptide concentration, 94.2% and 5.8% respec-
tively, exhibited hypocholesterolemic activity at varying
degrees in Wistar rats (Itou and Akahane 2009). In contrary,
effects of free amino acid mixtures and undigested blue
whiting protein tended to be negligible on cholecystokinin
(CCK) secretion from secretin tumor cell line (STC-1)
where its digested sarcoplasmic proteins significantly
increased the hormone level (+45% greater than the egg
albumin hydrolysate) (Cudennec et al. 2012). Interestingly,
among fractions peptide rich/free amino acid deficient
NH4OH-HCl fraction presented better hypocholesterolemic
activity on plasma lipid levels of rats than the peptide defi-
cient/free amino acid rich ACN fraction (Itou and Akahane
2009). Nonetheless, fecal lipids (bile acid (BA) and total
lipid (TL)) excretion was more affected by heshiko extract
(M/G 1.33) and its ACN fraction (M/G 1.16) compared to
NH4OH-HCl fraction (Itou and Akahane 2009). These
observations may point to the contribution of both free AA
and peptides to the anti-hypercholesterolemic activity of
the heshiko extract (Itou and Akahane 2009). The ACN
free amino acid rich fraction, with comparatively lower M/G
ratio of 1.16 compared to heshiko extract (M/G ratio 1.33),
was more modulatory, up to +29-times, on hepatic lipid
levels (TC: +13% (13 vs. 26%), LDL-c: +13% (13 vs. 26%),
TG: +29% (0 vs. 29%)) (Itou and Akahane 2009).
Marine biopeptides have exerted significant hypoglycemic,
insulinotropic, hypolipidemic, hypocholesterolemic, hypoten-
sive, pro-and anti-inflammatory and anti-obesogenic effects
in many studies thus being potential therapy candidates in
prevention and treatment risk factors (Ko and Jeon 2013;
Tørris, Småstuen, and Molin 2018). The topics of fish anti-
hypertensive and immunomodulatory biopeptides have been
carefully reviewed by the authors (Soheila Abachi, Bazinet,
and Beaulieu 2019; Soheila Abachi et al. 2021). Yet, herein
beneficial effects of similar bioactives on additional risk factors
of MetS are further comprehensively discussed. Many types
of fish and fish byproducts have cardio-protecting, anti-diabetes,
and anti-obesity potency yet interestingly fish diet itself has
also demonstrated health effects in many human intervention
studies (Tables 3–5). Nonetheless, marine, and fish-based
material or hydrolysates can be used as unprocessed diet,
dietary supplements and or as therapeutically active ingredi-
ents in food, nutraceutical, and pharmaceutical preparations
(Table 6). Fish biopeptides have proven to be safe and
non-cytotoxic thus being suitable alternatives to classic and
conventional drugs (Cudennec et al. 2008; Y. M. Kim etal.
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 11
Table 3. Fish peptides with bioactivities on MetS associated risk factors (in-vitro and cell studies).
Fish name and part used
Observed eect(s) including percentage of
eect and IC50
Characteristics of material and or peptide
(AA, sequence, and MW) Reference
Atlantic salmon (S. salar)
fresh skin byproducts
(recovered from skin-o
llets)
DPP-IV activity (5 mg non-hydrolyzed gelatin
and its hydrolysates mL-1 ≤ 45%, 2 mg mL-1
of UF fractions ≤ 61%,
100 g mL-1 of HPLC F1-F5 fractions ≤ 68%,
IC50: F1 57.3 g mL-1, GPAE 49.6 M, GPGA
41.9 M)
HAA 61 > C AA 24 > imino acids (Hyp + Pro)
18 > BCAA 4 mol 100-1 mol AA, L/A 0.47,
M/G 0.04, GPAE 372 Da, GPGA 300 Da
(Li-Chan et al. 2012)
Atlantic salmon (S. salar) skin
and trimmings byproduct
DPP-IV activity (IC50: skin lysates
0.90 − 2.13 mg mL-1, trimming lysates
0.84 − 1.27 mg mL-1),
High in Gly-Pro motif,
Skin: HAA 50 > CAA 27 > Gly 23 > Pro
11 > BCAA 5, L/A 0.38, M/G 0.11,
Trimming: CAA 34 > HAA 33 > BCAA 12 > Gly
7 > Pro 4, L/A 1.27, M/G 0.38
alcalase: <1 kDa (60 61%), 1 5 kDa
(35 − 38%),
alcalase + avourzyme: <1 kDa (67 69%),
1 − 5 kDa (28 − 32%),
promod: <1 kDa (33 59%), 1 5 kDa
(39 − 53%),
skin and trimming lysates digestion
susceptible and resistant respectively
(Harnedy et al. 2018a)
2.5 mg mL-1
intracellular cAMP concentration in
BRIN-BD11 cells (except skin lysates),
membrane potential (depolarization),
intracellular calcium ([Ca2+]i) concentration,
insulin secretion (insulinotropic response) from
BRIN-BD11 cells (except promod lysates),
GLP-1 secretion from enteroendocrine GLUTag
cells (except alcalase and promod lysate)
Barbel (B. callensis) fresh
muscle
DPP-IV activity (IC50: hydrolysate 1.94 mg
mL-1, SEC fraction 1.23 mg mL-1, RP-HPLC
fraction 0.21 mg mL-1, HPLC fraction 96 µg
mL-1)
WSG 330 Da, FSD 349 Da (Assaad Sila etal. 2016)
Barbel (B. callensis) fresh skin DPP-IV activity (IC50: lysates of esperase
2.2 mg mL-1, savinase 2.7 mg mL-1, alcalase
2.6 mg mL-1, trypsin 2.5 mg mL-1, izyme G
3.7 mg mL-1, protamex 2.4 mg mL-1,
neutrase 2.7 mg mL-1, peptidase 2.7 mg
mL-1)
HAA (HAA) 629 > Gly 354 > CAA 189 > imino
AA 184 > BCAA 45 residues/1000, L/A
0.52, M/G 0.04,
Lysate of esperase 2,935 Da, savinase
2,322 Da, alcalase 1,995 Da, trypsin
3,925 Da, izyme G 5,797 Da, protamex
2,325 Da, neutrase 7,878 Da, peptidase
15,053 Da
(Assaâd Sila et al. 2015)
Bighead carp (H. nobilis)
muscle
DPP-IV activity (lysates, fractions, and
sub-fractions (IC50 YNLKERYAAW 12.46 mM,
MKAVCFSL 1.37 mM, LGQNPAAML 0.96 mM,
INEFTTGIPVL 0.73 mM, IADHFL 0.61 mM),
and IC50 IPI (diprotin A) 0.005 mM
0.5 2 kDa (lysate of papain: 54%, alcalase:
76%, trypsin: 69%, pepsin: 82%),
MKAVCFSL 898 Da, YNLKERYAAW
1,314 Da, INEFTTGIPVL 1,203 Da, IADHFL
715 Da, LGQNPAAML 914 Da
(C. Zhang et al. 2017)
Blacktip shark (C. limbatus)
skin byproduct
LDL-c oxidation (≤ 39%), hydroxyl and
peroxyl radical-induced DNA scission
(Kittiphattanabawon
et al. 2013)
Blue whiting (M. poutassou)
fresh frozen muscle
0.5 − 1% (w/v)
CCK and GLP-1 in STC-1 cell
(dose-dependent)
(Cudennec et al. 2012)
Blue whiting (M. poutassou)
muscle
DPP-IV activity (IC50 1.28 mg mL-1), 1 − 5 kDa (21%), 0.5 − 1 kDa (29%), <0.5 kDa
(49%), C AA 26 > HAA 23 > BCAA 10 g
100 g-1, L/A 1.32, M/G 0.65
(Harnedy et al. 2018b)
2.5 mg mL-1
insulin secretion from BRIN-BD11 cells,
GLP-1 secretion from GLUTag cells,
insulin-stimulated glucose uptake in 3T3-L1
adipocytes
Boarsh (C. aper) muscle DPP-IV activity (IC50 1.18 mg mL-1), 2 1 kDa (18%), <1 kDa (74%) (Parthsarathy et al. 2019)
2.5 mg mL-1
insulin secretion from BRIN-BD11 cells,
GLP-1 secretion from GLUTag cells, glucose
uptake in 3T3L1 adipocytes (30%), [Ca2+]i,
membrane depolarization
Cod, commercial preparation
by DP&S®, Netherlands
GLP1 and CCK release (trypsin and or
DPPIV digested and or non-digested) in
STC1 cell
(Geraedts et al. 2011)
Fish scales, commercial
collagen preparation by
Nippi Co. Ltd., Japan
DPP-IV activity (IC50 3.5 mg mL-1, GA(HP) >
20 mM, GPA 5.03 mM, GP(HP) 2.51 Mm) and
IC50 IPI (diprotin A) 0.03 mM
GA(HP), GPA, GP(HP) (Hatanaka, Kawakami,
and Uraji 2014)
Herring milt hydrolysate
commercial preparation
1 ng mL-1
glucose uptake (27%) in insulin stimulated
L6 cells (except charged peptides and
non-electrodialyzed)
Neutral >50 kDa fraction: CAA 32 > EAA
15 > BCAA 7 > proline 3, L/A 0.13, M/G
0.37
(Durand et al. 2019)
Pacic hake (M. productus),
halibut (H. stenolepis),
tilapia (O. niloticus),
milksh (C. chanos) fresh
skins, byproducts
recovered from skin-o
llet
DPP-IV activity (3 mg hydrolysate mL-1: ≤
48%, and 1 mg UF fraction mL-1: ≤ 52%)
High in Gly, Pro and Hyp (tilapia and
milksh: 1.95 − 2.03 µmole imino acids
mg-1 sample, halibut and hake:
1.77–1.79 µmole imino acids mg-1
sample), halibut: SPGSSGPQGFTG 862 Da,
GPVGPAGNPGANGLN 1,021 Da,
PPGPTGPRGQPGNIGF 1261 Da,
tilapia: IPGDPGPPGPPGP 920 Da,
LPGERGRPGAPGP 1,027 Da,
GPKGDRGLPGPPGRDGM 1,359 Da
(T.-Y. Wang et al. 2015)
(Continued)
12 S. ABACHI ETAL.
Rainbow trout (O. mykiss)
fresh frames
DPP-IV activity (IC50: hydrolysate 4.8 mg
mL-1,
EDUF fraction (without cationic peptides)
2.1 mg mL-1, EDUF cationic peptides 1.2 mg
mL-1)
hydrolysate: HAA 47 > CAA 27 > BCAA 12%
mole, L/A 2.08, M/G 0.18, 0.5 1 kDa (
35%),
EDUF fraction (without cationic peptides):
HAA 41 > CAA 38 > BCAA 10% mole, L/A
1.96, M/G 0.17, 0.5 1 kDa ( 25%),
EDUF cationic peptides: CAA 33 > HAA
24 > BCAA 7% mole, L/A 2.23, M/G 0.21,
0.5 − 1 kDa ( 50%)
(Ketnawa et al. 2019)
Salmon (S. salar) and cod (G.
morhua) frozen byproduct
frames
1 mg mL-1
water-dispersed and or alkaline-solubilized
enzymatic lysates
insulin-induced glucose uptake in L6
myocytes,
glucose production in FAO rat hepatocytes
(except cod lysates)
(Jin 2013)
Salmon (S. salar) trimmings DPP-IV (IC50: hydrolysate 0.30 − 2.41 mg mL-1
and IC50: corolase hydrolysate-separated
peptides (GPAV 246 M, VP 758 M, VC
5413 M, YP 7564 M, FF 547 M, PP
4343 M, W 438 M, F 295 M, Y 75 M)
GI digestion stable corolase hydrolysate:
GPAV, VP, VC, YP, FF, PP, DP, I/LD, I/LH, W,
L/I, F, Y
(Neves et al. 2017)
Salmon frames 1 ng mL-1
glucose uptake ≤ 40% in insulin stimulated
and non-stimulated L6 skeletal muscle cells
CAA, <800 Da (90 − 92%) (Roblet et al. 2016)
Salmon fresh backbones 8 mg (papain + bromelain) mL-1
uptake of radiolabeled glucose (39%) into
CaCo-2 cells (cellular GLUT/SGLT ) activity)
papain + bromelain hydrolysate 250 − 300 Da (Slizyte et al. 2016)
Sardine (S. pilchardus), horse
mackerel (T.
mediterraneus), axillary
seabream (P. acarne),
bogue (B. boops),
small-spotted catshark (S.
canicula), blue whiting (M.
poutassou) west
Mediterranean Sea sh
discards
100 mg protein
BA (cholic and chenodeoxycholic acid)
binding capacity (sardine 13.1 25.0%,
horse mackerel 13.4 30.8%, axillary
seabream 17.7 − 20.9%, bogue 17.7 − 28.4%,
small-spotted catshark 14.1 17.9%, blue
whiting 24.3 28.2% relative to
cholestyramine)
(Pérez-Gálvez et al. 2015)
Siki (C. coelolepis) cooked
heads and saithe (P.
virens) cooked muscle
CGRP binding to receptors in rat liver
membrane (ED50: 1.68 mg protein (siki
lysate), 0.017 mg protein (siki lysate
puried fraction), 1.38 mg protein (saithe
lysate))
Siki hydrolysate: 800 − 1,500 Da (74%),
≤340 Da (26%), GFP(HP)GPEGL
(Martínez-Alvarez et al.
2012)
Silver carp (H. molitrix Val.)
fresh muscle
1.25 mg mL-1
DPPIV activity ≤ 57% (neutrase lysate and
fractions ≤ 56%) (IC50: APGPAGP
229.14 M, LPIIDI 105.44 M) and IC50 IPI
(diprotin A) 6.36 M
APGPAGP 566 Da, LPIIDI 683 Da (Ying Zhang, Chen, Chen,
et al. 2016)
Skipjack Tuna (K. Pelamis)
heart
PTP1B (IC50 416 g mL-1), αglucosidase
activity (IC50 1136 g mL-1), HRAR activity
(IC50 984 g mL-1)
(Ali et al. 2016)
Steelhead (O. mykiss) fresh
skin gelatin
0.5 mg mL-1
DPPIV activity ≤ 44%
(Cheung and Li-Chan
2017)
Tilapia (Oreochromis sp.)
scale, commercial food
grade collagen (Wellnex®
Type D) preparation by
Nitta Gelatin, Inc., Japan
DPP-IV activity (IC50 0.77 mg mL-1), (Iba etal. 2016)
1% protein
GLP-1 secretion (258%) in NCI-H716 cells
Tuna (T. tonggol) cooking
juice (5.44% protein)
DPP-IV activity (10 mg non-hydrolyzed
gelatin and its hydrolysates mL-1 ≤ 45%,
5 mg gel ltered fractions mL-1 ≤ 40%,
2 mg RP-HPLC fractions mL-1 ≤ 63%), IC50:
PGVGGPLGPIGPCYE 116.1 M, CAYQWQRPVDRIR
78.0 M, PACGGFWISGRPG 96.4 M
Protease XXIII lysate high in
PGVGGPLGPIGPCYE 1,413 Da,
CAYQWQRPVDRIR 1,691 Da,
and orientase lysate high in
PACGGFWISGRPG 1,305 Da
(S.-L. Huang et al. 2012)
Tuna extract 500 − 1000 ng mL-1 of DIVDKIEI (TP-D)
glucose uptake (preadipocytes
dierentiation), TG, adipocyte formation
(expression of C/EBP-α/-β/-δ and PPAR-γ),
expression levels of ACC, FAS, LPL, FABP,
SREBP-1, SOCS3, CD36, expression level of
mitochondrial UCP-2, GLUT4, and GSK-3β,
high-MW- and total adiponectin,
expression level of UCP-1, CCND1 and
β-catenin/TCF/LEF levels (Wnt-10b/LRP6/
Frizzled activated) in 3T3-L1 cells during
dierentiation
DIVDKIEI 944 Da (Y. M. Kim etal. 2015)
Table 3. (Continued).
Fish name and part used
Observed eect(s) including percentage of
eect and IC50
Characteristics of material and or peptide
(AA, sequence, and MW) Reference
(Continued)
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 13
Tuna skin byproduct 1 mg mL-1, 8-d
intracellular TG, lipid accumulation and lipid
droplets (adipogenic dierentiation of
preadipocytes), expression of adipogenic
genes (C/EBP-α and PPAR-γ) and target
genes (aP2) in 3T3-L1 preadipocytes,
(E. J. Lee et al. 2017)
1 mg mL-1, 24-h
palmitate-induced lipogenesis in HepG2
cells
Unicorn leatherjacket sh (A.
monoceros) byproduct
skin
α-amylase activity (IC50 hydrolysate (various
process temperatures) 5 °C 1.17 mg mL-1,
25 °C 1.92 mg mL-1, 50 °C 2.65 mg mL-1
(Kumar, Shakila, and
Jeyasekaran 2019)
Yellown tuna (T. albacares)
fresh bone
α-glucosidase activity ≤ 24% (Natsir, Dali, and Arif
2019)
Zebra blenny (S. basilisca)
muscle
α-amylase activity (IC50: 90 − 93 g mL−1)CAA 35% > BCAA 31% > sulfur containing
AA 3%, L/A 0.51
(Ktari et al. 2013)
Increased, improved, stimulated and or upregulated expressions are denoted as , and decreased, inhibited and or downregulated expression denoted as ”.
HAA: hydrophobic amino acids (glycine, alanine, valine, isoleucine, leucine, phenylalanine, proline, methionine, and tryptophan), AAA: aromatic amino acids
(phenylalanine, tryptophan, and tyrosine), PCAA: positively charged amino acids (arginine, histidine, lysine); NCAA: negatively charged amino acids (aspartic
acid, glutamic acid), EEA: essential amino acids (phenylalanine, valine, threonine, isoleucine, methionine, histidine, leucine, lysine and tryptophan), BCAAs:
branched chain amino acids (isoleucine, leucine and valine), TCF/LEF: T-cell factor/lymphoid enhancer factor, UCP-1 and UCP-2: Uncoupling protein a and 2,
GSK-3β: Glycogen synthase kinase 3 beta, SOCS3: Suppressor of cytokine signaling 3, ACC: AcetylCoA carboxylase, FAS: Fatty acid synthase, LPL: Lipoprotein
lipase, FABP: Fatty acid binding protein, SREBP-1: Sterol regulatory element binding protein-1c, SOCS-3: Suppressor of cytokine signaling-3, CD36: Cluster of
dierentiation 36, GLUT4: Glucose transporter type 4, GSK-3β: Glycogen synthase kinase-3β, CCND1: Cyclin D1, TCF/LEF: T-cell factor/lymphoid enhancer factor,
LRP6: Low-density lipoprotein receptor-related protein 6, Wnt-10b: Wingless-type MMTV integration site family, GLUT: Glucose transporter, SGLT: Sodium
glucose cotransporter.
Table 3. (Continued).
Fish name and part used
Observed eect(s) including percentage of
eect and IC50
Characteristics of material and or peptide
(AA, sequence, and MW) Reference
2015; E. J. Lee et al. 2017; Nasri et al. 2015). Anti-diabetic
fish biopeptides exert their effect through different mecha-
nisms nevertheless they commonly lower blood glucose and
activities of α-amylase, α-glucosidase, and DPP-IV, increase
intracellular calcium concentration, insulin, CCK and GLP-1
secretion, glucose uptake and clearance improving glucose
tolerance and preventing insulin resistance. Like glucoregula-
tory fish biopeptides, hypolipidemic, anti-obesogenic and
anti-MetS peptides act via various modes too but generally
by modulating plasma and hepatic lipid parameters, lipid
peroxidation, bile acid binding and excretion, food intake and
body weight gain (lean and fat), expression of adipogenic
genes, appetite related proteins and their expression, athero-
genic index (AI) and coronary risk index (CRI). Not only
that fish anti-MetS peptides have been fairly stable in the
gastrointestinal (GI) environment but also proteolytic activity
of the digestive tract in many examples have shown to improve
the bioactivity of these compounds. Moreover, many of the
fish anti-hyperglycemic and or anti-hyperlipidemic peptides,
based on their absorption, distribution, metabolism, and excre-
tion (ADME) properties, are druglike compounds which could
further be used on as pharmaceutical biomolecules. In the
following text, authors will discuss the topics of production
of anti-MetS fish biopeptides, effects of these products on
metabolic impairments, their multi-functional properties and
structure-activity relationship including their susceptibility to
GI proteases.
Extraction, isolation, and purication techniques
A broad range of enzymes are in use for the preparation
of anti-MetS biopeptides from fish and fish products or
byproducts. Enzyme choices range from pure or mixed
commercial enzymes to alkaline, neutral, acid proteases and
or in-house prepared crude plant-, fish- and or
bacterial-proteases. Fish enzymes were derived from the
digestive tract of many types of fish such as zebra blenny,
smooth hound, etc. Typically, bacterial crude enzymes were
derived from Bacillus species, yet handful studies have made
use of Aspergillus and or Streptomyces species. Plant-originated
enzyme was prepared from papaya latex for hydrolysis of
shark skin (Kittiphattanabawon etal. 2013). Among the so
commonly used enzymes some peculiar ones e.g., seabzyme,
izyme, etc. are also found in literature. It is of note that
enzymatic hydrolysis has not always been the best course
for preparation of glucoregulatory fish biopeptides since it
could under certain experimental conditions diminish and
or interrupt the bioactivity (Li-Chan etal. 2012; T.-Y. Wang
et al. 2015). For efficient isolation, filtration methods were
applied to the material in the majority of the studies.
Furthermore, except in a few studies for selective separation
and purification, as routine for fish hypotensive and immu-
noregulatory peptides, chromatographic procedures were
commonly practiced (Ktari et al. 2013; Nasri et al. 2018;
Nasri et al. 2015; Siala et al. 2016).
Antidiabetic fish peptides were generally prepared by enzy-
matic hydrolysates however to optimize the digestion various
studies employed pretreatments of some sort like microwave
and alkaline/acidic solubilization particularly those attempting
to isolate gelatinous and or collagenous material out of fish
byproducts. Handful studies effectively proceeded without
hydrolyzing the material (e.g., preparation of soluble and
insoluble proteinous fractions) (Kato etal. 2011; Y. M. Kim
et al. 2015; Madani et al. 2012). Among many enzyme
choices, flavourzyme has been superior over alcalase,
14 S. ABACHI ETAL.
Table 4. Fish peptides with bioactivities on MetS associated risk factors (in-vivo studies with mono- and or multi-functionalities).
Fish name and part used Observed eect(s)
Characteristics of material and or peptide
(AA, sequence, and MW) Reference
In-vivo studies (mono-functional)
Alaska pollack (T.
chalcogramma) llet
100 mg kg−1 (intraperitoneal)
blood glucose in T2D KK-Ay rats,
ANGEVAQWR 1,029 Da, IWHHTFYNELR
1,515 Da
(Ayabe etal.
2015)
3 mg kg−1 ANGEVAQWR (intraperitoneal)
blood glucose in T2D KK-Ay rats,
1 mg kg−1 of QWR (intraperitoneal)
blood glucose in T2D NSY and KK-Ay rats
Alaska pollack (T.
chalcogramma) llet
300 mg kg−1 BW, 2-times daily, 2- and 3-d
(intraperitoneal)
total food intake and total BW gain, perirenal and
epididymal adipose tissue weight, sum of perirenal
and epididymal adipose tissue weights, appetite
related mRNA expression in hypothalamus
(neuropeptide Y), agouti-related protein,
soleus skeletal muscle weight in rats
(Mizushige et al.
2017)
Alaska pollack (T.
chalcogramma) meat,
commercial
water-insoluble
dehydrated protein
preparation
197 g kg-1 diet (methionine supplemented (low- (2.5 g
kg-1 diet) and high-Met (5 g kg-1 diet)), ad libitum,
28-d
plasma cholesterol,
mRNA levels of IBAT, ApoB, SREBP-1a, cholesterol
12α-hydroxylase and fecal BA (except high-Met
diet), BA in small-intestinal contents, mRNA levels of
SREBP-1c in OVX rats
CAA 84 > HAA 57 > BCAA 31 g kg-1 diet, L/A
2.0, M/G 0.38
(Kato et al. 2011)
Alaska pollock (T.
chalcogramma) llet
10% sh protein or 2% pure tuna oil (FO) or sh
protein + FO, 4-wks
serum TG (≤ 17%), cholesterol (≤ 18% except FO),
and LDL-c (≤ 8%), hepatic TG (≤ 50% except sh
protein), and cholesterol (≤ 32% except FO),
HDL-c (≤ 9% except FO), fecal cholesterol (≤ 32%
except FO), and BA (≤ 74%) in rats
CAA 50 > HAA 42 > BCAA 19 > AAA 9 g 100 g-1
protein, L/A 1.56, M/G 0.92
(Hosomi et al.
2013)
Alaska pollock (T.
chalcogramma) llet
4-wks diet
serum cholesterol (15%), TG (15%), and LDL-c (26%),
hepatic cholesterol (27%),
serum HDL-c (11%), fecal cholesterol and BA (102%)
compared to casein in hypercholesterolemic diet fed
rats
HAA 430 > C AA 420 > BCAA 187 g kg-1
protein, L/A 1.82, M/G 0.45
(Hosomi et al.
2012)
Alaska pollock (T.
chalcogramma) llet
10% casein + 10% sh (100 g kg-1 diet) protein (w/w),
4-wks
serum cholesterol and LDLc, hepatic cholesterol,
fecal cholesterol, BA acid, and nitrogen excretion
compared to 20% casein diet in
hypercholesterolemic fed rats
CAA 47 > HAA 39 > BCAA 18 g 100 g-1 protein,
L/A 1.56, M/G 0.92
(Hosomi et al.
2011)
Alaska pollock (T.
chalcogramma) llet
113.8 g kg-1 diet, 4-wks
serum TG (42%), NEFA (29%), AST (18%), and ALT
(20%), hepatic TL (43%), and TG (29%), relative
mRNA SCD-1 expression level (59%), hepatic FA and
FA desaturase indices
fecal FA (37%) compared to casein in obese T2D
KK-Ay rats
High in myobril proteins (myosin heavy
chain 120 kDa and actin 45 kDa), CAA
471 > BCAA 184 > AAA 87 > Gly 36 > Pro
33 g kg-1 total AA, L/A 1.56, M/G 0.92
(Maeda et al.
2015)
Allaska pollock (T.
chalcogramma) llet
4-wks diet
serum cholesterol (≤ 18%), serum LDL-c (≤ 17%),
liver cholesterol (≤ 32%),
serum HDL-c (7%) compared to casein in
hypercholesterolemic diet fed rats
(Hosomi et al.
2010)
Bester sturgeons (Huso
huso × Acipenser
ruthenus) skin, n, and
bone
1.5 g kg-1 BW, single oral injection
blood glucose levels in rats
Atelocollagen (type I collagen): βchain
220 kDa, collagen fraction: A(HP)
GPAGPTGK 868 Da, A(HP)GPVGPAGP
835 Da, E(HP)GPAGP(HP)GP 907 Da,
TGGIGG(HP)GGS 775 Da, AVGPVGPIGP
863 Da, AAGPHPG 485 Da, A(HP)GPAG
485 Da, GPGGPA 455 Da, L(HP)GPTG
557 Da, GPLGPA 511 Da, APAG 314 Da,
GYGPGPA 618 Da, GGYE 424 Da, GEYGP
522 Da, F(HP)GG(HP)GAKG 819 Da, FVA
335 Da, APNPFRHY 1,001 Da
(Sasaoka et al.
2018)
Blue whiting (M.
poutassou) muscle
100 mg kg-1 BW
blood glucose,
insulin release in normal rats
1 − 5 kDa (21%), 0.5 − 1 kDa (29%), <0.5 kDa
(49%),
CAA 26 > HAA 23 > BCAA 10 g 100 g-1, L/A
1.32, M/G 0.65
(Harnedy et al.
2018b)
Boarsh (C. aper) muscle 50 mg kg-1 BW
insulinotropic response, glucose tolerance in oral
glucose challenged normal rats
2 1 kDa (18%), <1 kDa (74%) (Parthsarathy
et al. 2019)
(Continued)
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 15
Chum salmon (O. keta)
and rainbow trout (O.
mykiss) skin,
commercial > 90%
purity collagen
preparation)
0.4 g single dose (oral)
plasma TG in fat fed rats,
3 − 10 kDa
HAA 63% > Gly 37% > CAA 23% > imino
acid 15% > BCAA 45%
(Saito et al. 2009)
0.17% sh collagen, 14-d
plasma TL, TG and VLDL-TG in fat fed rats
Cod llet powder (93%
protein)
21 g d-1 (19.61% protein) diet, 21-d
BW gain pre-injury (6%), sham muscle mass at 3-d
post-injury, sham (13%) and injured muscle (12%)
mass at 14-d post-injury, myober cross-sectional
area in sham muscle at 14-d post-injury compared
to casein in rats
EAA 43 > C AA 47 > BCAA 19 g 100 g-1 AA, L/A
1.83, M/G 0.82
(Dort et al. 2012)
Pacic hake (M.
productus), halibut (H.
stenolepis), tilapia (O.
niloticus), milksh (C.
chanos) fresh skins,
byproducts recovered
from skin-o llet
750 mg kg-1 d-1 28-d
blood glucose (except pacic hake and milksh),
DPP-IV activity (except pacic hake, halibut, and
milksh) 28%,
total GLP-1 and secretion (except pacic hake and
milksh) in streptozotocin-induced diabetic rats
High in Gly, Pro and Hyp (tilapia and
milksh: 1.95 − 2.03 µmole imino acids
mg-1 sample, halibut and hake;
1.77 − 1.79 µmole imino acids mg-1
sample), halibut: SPGSSGPQGFTG 862 Da,
GPVGPAGNPGANGLN 1,021 Da,
PPGPTGPRGQPGNIGF 1261 Da, tilapia:
IPGDPGPPGPPGP 920 Da, LPGERGRPGAPGP
1,027 Da, GPKGDRGLPGPPGRDGM 1,359 Da
(T.-Y. Wang et al.
2015)
Salmon (S. salar) and cod
(G. morhua) frozen
byproduct frames
10% salmon (<1 kDa) protein, 3-mos
fasting hyperinsulinemia and glucose intolerance,
hepatic glucose production from gluconeogenesis in
HFHS fed LDLR-/-/ApoB100/100 rats
(Jin 2013)
Sardine (S. pilchardus) and
bogue (B. boops) fresh
muscle
1 g kg-1 BW d-1, 30-d
serum lipids (TC 66%, TG ≤ 41%, PL ≤ 53%,
unesteried cholesterol ≤ 36%, cholesteryl ester ≤
68%), liver lipids (TC ≤ 22%, cholesteryl esters ≤
29%, unesteried cholesterol ≤ 15%),
serum apolipoprotein A-IV ≤ 33%, fecal cholesterol
excretion in hypercholesterolemic diet fed rats
CAA 34 36% > BCAA 9 20% > sulfur
containing AA 7 − 14%
(Benomar et al.
2015)
Sardine (S. pilchardus) and
sardinelle (S. aurita)
fresh muscle
300 mg sh diet, 14-d (oral gavage)
serum lipid parameters (TC ≤ 42%, TG ≤ 46%,
unesteried cholesterol ≤ 35%, cholesteryl ester ≤
51%), serum LDL-HDL1 ≤ 57%, AI ≤ 17%,
HDL2 ≤ 22%, HDL3 ≤ 40% in hypercholesterolemic
diet fed rats
(Athmani et al.
2015)
Siki (C. coelolepis) cooked
heads and saithe (P.
virens) cooked muscle
50 mg d-1 per rat, 21-d
plasma CGRP levels (70%) (except saithe lysate)
Siki hydrolysate: 800 − 1,500 Da (74%),
≤340 Da (26%), GFP(HP)GPEGL
(Martínez-Alvarez
et al. 2012)
Smooth hound (M.
mustelus) muscle
5 mg d-1 per rat, 3-wks (oral)
BW gain, food intake in rats
CAA 30% > BCAA 21% > Gly 12% > Ala
10%, L/A 1.46
(Bougatef et al.
2010)
Tilapia (Oreochromis sp.)
scale, commercial food
grade collagen
(Wellnex® Type D)
preparation by Nitta
Gelatin, Inc., Japan
3 g kg-1
glycemic response during OGTT (28%) and IPGTT
(18%), intestinal glucose uptake
glucose-stimulated active GLP-1 and insulin secretion
in normal rats
(Iba et al. 2016)
Tuna skin byproduct 300 mg kg BW d-1, 8-wks (oral)
BW gain, expression of adipogenic genes and
transcription factors, epididymal adipocyte size,
plasma TC (11%), TG (6%), and LDL-c (14%),
HDL-c (19%) in HFD-fed obese rats
(E. J. Lee et al.
2017)
Zebra blenny (Salaria
basilisca) fresh muscle
400 mg kg-1 BW d-1, 8-wks (gastric gavage)
serum AST and LDH activities, lipid peroxidation,
protein oxidation and ROS production, TBARS, AOPP
and H2O2 in heart tissues, GPx, SOD, and CAT,
ATPase activities and GSH in heart tissues back to
normal levels in hypercholesterolemic diet (1%
cholesterol + 0.1% cholic acid) fed rats
Zebra blenny, smooth hound, and sardinelle
proteases: 5 20 kDa (46, 49, 49%),
<5 kDa (43, 42, 27%)
(Ktari et al. 2017)
In-vivo studies (bi- and or multi-functional)
Atlantic herring (C.
harengus) byproducts
(heads, guts, and
backbones), Atlantic
salmon (Salmo salar)
byproducts (backbones)
5% sh hydrolysate + 15% casein/whey, 4-wks
daily dietary intake of sodium, chloride, and
potassium,
urine concentrations (relative to creatinine
concentration) of total protein, glucose and cystatin
C, urine concentrations of potassium
(Drotningsvik,
Pampanin,
et al. 2018)
Atlantic salmon (S. salar)
fresh skin byproducts
(recovered from skin-o
llets)
300 mg d-1, 5-wks
blood glucose levels, plasma DPP-IV activity, food,
and water intake,
active GLP-1 secretion, insulin, insulin-to-glucagon
ratio, body weight in STZ-induced diabetic rats
(Hsieh et al.
2015)
Table 4. (Continued).
Fish name and part used Observed eect(s)
Characteristics of material and or peptide
(AA, sequence, and MW) Reference
(Continued)
16 S. ABACHI ETAL.
Atlantic salmon (S. salar)
frozen frames
10.75 g peptides kg-1 diet, 12-wks
body and liver weight, feed eciency, plasma TG
and glycerol, glucose intolerance in LDLR−/−/
ApoB100/100 rats
EAAs 46 > HAA 43 > CAA 40 > BCAA 19 g
100 g −1 protein
(Chevrier et al.
2015)
Blue whiting (M.
poutassou) fresh frozen
muscle
100 − 250 mg, 12- d (oral)
food consumption, weight gain (except 250 mg),
active GLP-1 (≤ 55%), plasma CCK (≤ 66%) (dose
dependent) in rats
(Cudennec et al.
2012)
Blue whiting fresh frozen
whole
33% sh (108 g kg-1 diet) + 67% casein, 5-wks
serum TC, cholesteryl ester, LDL-c and HDL-c, hepatic
cholesterol, HMG-CoA reductase, and LDL receptors
compared to casein obese Zucker fa/fa rats
HAA 59 > BC AA 34 > PCAA 27 > Tau 0.2 g kg-1
diet, L/A 2.08, M/G 0.65, >20 kDa 37%,
10 − 20 kDa 13%, 0.2 − 10 kDa 13%,
<0.2 Da 37%, GGV
(Drotningsvik,
Vikøren, et al.
2018)
Bogue (B. boops) llet 0.1, 0.5 and 2 g kg-1 BW d-1 (digested and
non-digested)
TC (≤ 35%), TG (≤ 43%), LDL-c (≤ 72%), ALT, AST,
atherosclerotic plaques (including formation of foam
cells),
HDL-c (≤ 22%) in hypercholesterolemic diet fed rats
HAA 45 − 48%, CAA 28 − 29%, BCAA
14 − 14%, AAA 8 − 9%, L/A 0.23 − 0.33,
M/G 0.22 − 0.26
(Lassoued et al.
2014)
Bonito, herring, mackerel,
salmon, commercial
preparations by Ocean
Nutrition (Nova Scotia,
Canada), Aquatic
Products Technology
Center (Quebec,
Canada), Marine Harvest
Ingredients (Hjelmeland,
Norway)
20% protein (w/w), 28-d
BW gain (gross weight gain eciency ratio), dietary
fat mediated accretion of eWAT (visceral adiposity)
(except bonito, herring, mackerel),
energy expenditure, glucose infusion rate, circulating
sCT (except bonito, herring, mackerel) in HFHS (20%
lard + 22% sucrose + 0.2% choline bitartrate) diet
fed rats
(Pilon et al. 2011)
Chub mackerel (fermented),
commercial preparation
(51.1% crude protein)
10 − 20 g kg-1 diet
ACC, FAS, serum TG, AI,
feed intake, body weight gain, CYP7A1, HDL-c in
diabetic rats
(Santoso,
Ishikawa, and
Tanaka 2010)
Cod (T. chalcogramma)
and tuna (T. orientalis)
light muscle
23% protein diets, 4-wks
weight gain, nal weight, food intake, fecal acidic
sterols, and nitrogen (except cod), fecal neutral
sterols and HMGCR mRNA expression (except tuna),
SHP-1 mRNA expression,
serum cholesterol, HDL-c, non-HDL-c, PL, NEFA,
relative liver weight, and hepatic cholesterol (except
tuna), CYP7A1 mRNA expression compared to casein
in hypercholesterolemic diet fed rats
myosin 116 kDa and actin 45 kDa,
Cod: CAA 47% > HAA 39% > BCAA 16%,
L/A 1.17, M/G 0.71,
Tuna: CAA 47% > HAA 38% > BCAA 16%,
L/A 0.99, M/G 0.79
(Hosomi et al.
2017)
Cod llet 200 g protein kg-1 diet for low fat and or 400 g protein
kg-1 diet for high-fat/high-protein, 6- and 12-wks
(ad libitum)
further weight and fat mass gain, feed eciency,
lean mass, insulin sensitivity in hyper- and
normo-caloric diet fed C57BL/6J (obese or lean) rats
(Myrmel et al.
2019)
Cod powder, commercial
non-hydrolyzed
preparation by
Seagarden AS, Norway
5% cod + 15% casein/whey + (w/w), 4-wks (ad
libitum)
BW and growth percentage, liver weight, cholesterol
level in liver,
postprandial blood glucose in obese Zucker fa/fa rats
(Drotningsvik
et al. 2015)
Fish collagen, commercial
preparation by Nippi
Inc., Japan and Oriental
Yeast Co. Ltd., Japan
6% casein + 4% sh collagen, 10-wks
food intake, hepatic expressions of lipid metabolism
involved genes and their targets,
adiponectin, TC, free cholesterol, esteried
cholesterol, expression of unfolded protein response
involved genes compared to casein in rats
MW 4 − 6 kDa (Tometsuka et al.
2017)
Fresh Norwegian Atlantic
salmon o-cut spine
byproducts, commercial
91% pure protein
preparation by Marine
Bioproducts AS, Norway
5% (w/w), 12-wks
plasma non-esteried FA, C18:2n-6 (linoleic acid) and
C20:4n-6 (arachidonic acid)
plaque area in sinus and aortic arch, hepatic mRNA
level of ACACA, SCD1 compared to casein diet in
ApoE-/- rats
<1,200 Da (<200 Da 25%) (Parolini et al.
2014)
Fresh Norwegian Atlantic
salmon o-cut spine
byproducts, commercial
91% pure protein
preparation by Marine
Bioproducts AS, Norway
165 g kg-1 diet (15% (w/w)), 2-wks
hepatic activity of FA synthase,
weight gain, feed intake, hepatic activities of
peroxisomal ACOX1 and mitochondrial carnitine
palmitoyltransferase-II in hTNF-α mice
(Bjørndal et al.
2013)
Table 4. (Continued).
Fish name and part used Observed eect(s)
Characteristics of material and or peptide
(AA, sequence, and MW) Reference
(Continued)
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 17
Goby sh (Z.
ophiocephalus) fresh
muscle
400 mg kg-1 BW d-1, 10-wks (gastric gavage)
plasma glucose 25 34%, α–amylase activity ≤ 62%,
hepatic glycogen, hepatic and renal MDA levels,
creatinine, renal damage (improvement of kidney
architecture)
hepatic and renal GPx, CAT, SOD and GSH activities,
uric acid in hypercaloric (high fat high fructose: 10%
animal fat + 5% fructose + 0.1% cholic acid) diet
fed rats
HAA 44 47% > EAA 39 42% > CAA
27 30% > Gly 15 16% > BCAA
13 − 15%, L/A 0.46 − 0.58, M/G 0.13 − 0.19
(Nasri et al. 2015)
Goby sh (Z.
ophiocephalus) fresh
muscle
400 mg kg-1 BW d-1, 10-wks (gastric gavage)
weight gain, food intake ≤ 38%, TC, TG, LDL-c and
VLDL-c levels, AI, AIP and CRI, hepatic TG ≤ 43%
and TC ≤ 47%, pancreatic lipase activity ≤ 45%,
liver structure damage, lipid accumulation
HDL-c in high fat/ fructose diet rats
EAA 42% > HAA 40% > CAA 27% >
BCAA 14% > AAA 8%, sulfur
containing AA 5%,
(Nasri et al. 2018)
Gray triggersh (B.
capriscus) muscle
TG, TC, LDL-c, serum α -amylase activity, blood
glucose, hepatic, and serum GHbA1c, serum
bilirubin, lipid accumulation in hepatocytes
(pancreatic β-cells degeneration), ALT, ALP, and GGT
activities in hepatic and pancreatic tissues
HDL-c in alloxan-induced diabetic rats
High Lys and Arg (Siala et al. 2016)
Mixture of wild caught cod
lets and Canadian
scallop muscles (cod/
scallop, 1 : 1 on amino
acid content)
13-wks diet (ad-libitum)
weight gain, atherosclerotic plaque burden, leptin,
hepatic fat and fatty acid concentration, serum
LDL-c and glucose levels, hepatic gene expression of
antioxidative PON2 and VCAM1 in ApoE−/− rat
CAA 62 > HAA 58 > EAA 56 > BCAA 23 mg g-1
diet, L/A 1.48, M/G 0.62
(I.-J. Jensen et al.
2016)
Mixture of wild caught cod
llet and Canadian
scallop muscles (cod/
scallop, 1 : 1 on amino
acid content)
200 g protein kg-1 diet, 7-wks (pair-feeding design)
body mass gain, adipose tissue masse, hepatic TG,
glucose clearance, energy expenditure and activity in
obesity prone C57BL/6J mice
CAA 550 > EAA 512 > BCAA 237 > Gly
192 > sulfur containing AA 137 > Tau
61 mmol/kg, L/A 1.59, M/G 0.18
(Tastesen et al.
2014)
Nile tilapia (O. niloticus)
fresh
skin
1.7 g kg−1 BW, 25-d
typical symptoms of T2D (hunger and thirst), weight
gain, blood glucose levels (≤ 32%) in alloxan
induced diabetic rats
CAA 23 > imino acids 21 > Gly 19 g 100-1 g,
<1,000 Da 77% (short peptides < 9 AAs),
L/A 0.05, M/G 0.07
(R. Zhang, Chen,
Chen, et al.
2016)
Norwegian spring
spawning herring and
salmon (backbones)
fresh byproducts
114 g kg-1 diet (25% sh lysate + 75% casein/whey),
4-wks
serum LDL-c, HDL-c, TC, cholesteryl ester levels, total
serum n-6 PUFA (except salmon), postprandial
glucose, WAT 16:0 and total SFA (except herring),
20:4n-6,
mean growth %, serum 18:2n-6, WAT 18:3n-3 and
total n-3 PUFA (except herring), serum TG, -9
desaturated MUFA (16:1n-7 and 18:1n-9), total
MUFA, 22:5n-3, n-3:n-6 PUFA ratio (except salmon),
18:3n-3 compared to casein/whey in obese Zucker
fa/fa rats
<4 kDa (93 − 98%), 200 − 500 Da (14%),
200 − 1,000 Da (31 − 32%), <200 Da
(herring: 34%, salmon: 20%),
Herring: GPL, IPI, VW, GPAE, LGPG, IIAEK,
PGPL, HAA 66 > BCAA 41 > PCAA 35 > Tau
1 g kg-1 diet, L/A 1.6, M/G 1.0,
Salmon: GPL, IPI, VW, PGPL, HAA 68 > BCAA
42 > PCAA 32 > Tau 0.4 g kg-1 diet, L/A 2.0,
M/G 0.9
(Drotningsvik
et al. 2016)
Pollack (T. chalcogramma)
frame byproduct
11, 16, 21 and 26% of 310 g kg−1 sh meal (w/w) +
690 g kg−1 plant protein), hand-fed to apparent
satiation 2-times daily, 2-wks
Liver IGFI mRNA level
nal BW and SGR, feeding rate in high plant protein
fed Japanese ounder (Paralichthys olivaceus)
< 1,000 Da (93%), non-EAA 176 > CAA
166 > EAA 151 > BCAA 54 g kg−1 dry diet,
L/A 0.97, M/G 0.32
(Zheng et al.
2014)
Pollack (T. chalcogramma)
frame byproduct
6.2 12.4% of sh meal (w/w) (10 20% of diet’s total
protein + plant protein) 12-wks (hand-fed to
apparent satiation 2-times daily)
SGR, FER, PER, PR, VSI, serum TG, TC and LDL-c,
food intake in high plant protein fed Juvenile turbot
(Scophthalmus maximus)
>2 kDa (1%), 0.5 − 2 kDa (27%), 0.2 − 0.5 kDa
(57%), <0.2 Da (15%), non-EAA 20% >
EAA 18%, CAA 18% > BCAA 7%, Tau
0.15% dry matter, L/A 1.19, M/G 0.50
(Xu et al. 2016)
Pollock (T. chalcogramma)
esh
Hydrolysate and UF fractions 40% of sh meal diet,
30-d (hand-fed to apparent satiation 5-times daily)
mRNA expression of peptide transporter 1 (PepT1)
and CCK,
AST and ALT activities (except hydrolysate) in large
yellow croaker (Larimichthys crocea) larvae
Hydrolysate: (2 − 0.5 kDa 59%, < 0.5 kDa
41%), UF retentate: (2 0.5 kDa 76%,
<0.5 kDa 24%), UF permeate: (2 − 0.5 kDa
25%, <0.5 kDa 74%), EAA 35, non-EAA
35 > CAA 33 > BCAA 13 g 100 g-1 dry
matter, L/A 1.47, M/G 0.64
(Cai et al. 2015)
Table 4. (Continued).
Fish name and part used Observed eect(s)
Characteristics of material and or peptide
(AA, sequence, and MW) Reference
(Continued)
18 S. ABACHI ETAL.
Salmon spine and or
backbones including
heads, commercial
preparation by Marine
Bioproducts AS, Norway
(15% casein + 5% salmon peptide), 6-wks
BW gain, total weight gain, plasma and hepatic TG,
FAS activity, hepatic MUFA, FADS1 and HMGCR
expression (except umamizyme spine lysate), liver
index (% liver weight/BW) and hepatic TC (except
spine lysates), hepatic ACACA and FADS2 expression
(except umamizyme spine and alcalase backbone
lysates), n-3 and n-6 PUFAs (except acid protease A
spine and alcalase backbone lysates),
feed intake, FAS activity, hepatic n-6 PUFAs (except
umamizyme spine and alcalase backbone lysates),
plasma TG, TC, PL, HDL-c and hepatic 9 desaturase
mRNA level, 9 desaturase index and MUFA (except
acid protease A spine and alcalase backbone
lysates), hepatic n-3 PUFAs, NEFA, β-oxidation, 5
and 6 desaturase (n-3) index (except spine lysates),
hepatic ACOT1 expression (except acid protease A
spine lysate), hepatic 5 desaturase (n-6) index, and
PL (except umamizyme spine lysate) compared to
20% casein in high fat fed rats
Spine hydrolysates: 200 − 1,200 Da (>50%),
<200 Da (25%), backbone and head
hydrolysate: <1,200 Da (60%), HAA
42 48% > CAA 38 44% > BCAA
17 − 23% > imino AA 10 − 13%
(Vik et al. 2015)
Sardine (C. pilchardus)20% protein (w/w), 28-d
serum albumin, relative liver weight, RBC MDA and
SOD activity, epididymal fat and heart CAT activity
compared to casein in hypercholesterolemic diet
(1.5% cholesterol + 0.75% cholic acid) fed rats
(HamzaReguig
et al. 2013)
Sardine (S. pilchardus)
fresh byproducts
(viscera, heads, skins,
and edges) and llets
20% (200 g kg-1 diet) llet and/or byproducts protein,
4-wks
BW gain 41 84%, food intake 19 36%, food
eciency rate 27 75%, nal BW ≤ 6%, liver
relative weight ≤ 11%, adipose tissue relative
weight ≤ 18%, lipid intake 19 36%, serum leptin
14 − 29% and LDL-HDL1-c 43 − 56%, serum TC
27 − 31%, VLDL-c 55 − 62%, VLDL-TG 31 − 41%, liver
TG 29 − 37%, HDL3-PL 43 − 48%, HDL3-UC 26 − 35%,
serum and lipoproteins TBARS ≤ 64%,
fecal lipid 21 35%, and cholesterol 27 47%,
plasma HDL2-c 50 62%, and HDL3-c 25 − 35%, LCAT
activity 35 − 57%, HDL3-apo 19 − 49%, HDL2-CE
88 − 108%, serum and lipoproteins PON-1 ≤ 119%
compared to casein in high fat fed obese rats
Byproduct protein diet: CAA 43% > HAA
38% > BCAA 18% > Gly 6% > Tau 5%,
L/A 1.33, M/G 0.51
(Aane et al.
2018)
Sardine llet (head,
internal organs and
bones excluded)
200 g kg-1 diet (with or without fructose), 8-wks
food intake, perirenal and epididymal adipose tissue
wet weight, body composition index, plasma
glucose, insulin, TG, FFA, brinogen, HOMA-IR index
and HbA1c, perirenal and brown adipose tissues
hydroperoxide,
plasma α-tocopherol, taurine and calcium, brown
adipose tissue GSH-Px activity, epididymal and
perirenal WAT SOD activity, perirenal, epididymal
and BAT CAT activity compared to casein in high
fructose diet fed rats
(Madani et al.
2012)
Sardine muscle 1 g kg-1 BW d-1, 4-wks (oral)
plasma glucose (13%), heart weight and ratio of
heart weight to BW, urinary albumin excretion
(73%) in stroke prone SHR at 4 weeks
(Otani et al.
2009)
Sardine muscle tissue 20% sh diet, 8-wks
plasma glucose, insulin, creatinine, uric acid, and
homeostasis model assessment-insulin resistance
index levels, weight gain, food, and energy intakes
GLP-1 secretion in high fructose fed rats
CAA 45 > HAA 41 g 100 g-1 protein (Madani et al.
2015)
Sardinelle (S. aurita) fresh
muscle
400 mg kg−1 BW, 10-wks (gastric gavage)
fat accumulation and epididymal fats ≤ 31%, serum
TC ≤ 40%, TG ≤ 23.5%, LDL-c 43% and
VLDL-c 40%, atherogenic index, atherogenic index
of plasma ≤ 60% and coronary risk index ≤ 45%,
hepatic TG ≤ 38% and TC ≤ 53% concentrations,
lipase activity, liver fatty inltration and formation
of lipid vacuoles, hypertrophy of cells in infarction
of heart tissue, lipid deposition within intima of
aorta and lesion development in aortic wall,
HDL-c in hypercaloric diet fed rats
High HAA, MW 150 900 Da (B. subtilis A26
(150 − 600 Da) > B. amyloliquefaciens
An6)
(Jemil, Abdelhedi,
et al. 2017)
Table 4. (Continued).
Fish name and part used Observed eect(s)
Characteristics of material and or peptide
(AA, sequence, and MW) Reference
(Continued)
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 19
Sardinelle (S. aurita) fresh
muscle
400 g kg − 1 BW d-1, 10-wks (gastric gavage)
plasma glucose ≤ 22%, α-amylase activity ≤ 45%,
hepatic glycogenesis ≤ 71%, hepatic and renal MDA
levels, hepatic ALP ≤ 25%, renal creatinine 45%,
renal glomerular atrophia, hepatic tissue damage
hepatic and renal SOD activity (≤ 52% and ≤ 42%),
hepatic and renal GPx activity (≤ 32% and ≤ 84%),
hepatic and renal CAT activity (≤ 41% and ≤ 62%),
hepatic and renal GSH activity (≤ 30% and ≤ 41%)
in hypercaloric diet (10% sheep fat, 5% fructose,
0.1% cholic acid) induced hyperglycemic and
oxidative stressed rats
B. subtilis A26 product: EAA 45 > CAA 27,
HAA 27 > imino acids 5, BCAA 5 > Tau 0.4
residue 100-1 AA residues, L/A 2.71, M/G
0.55,
B. amyloliquefaciens An6 product: EAA
40 > CAA 36, HAA 21 > imino acids
5 > BCAA 2 > Tau 1.8 residue 100-1 AA
residues, L/A 2.37, M/G 0.38,
undigested product: EAA 36, CAA 35 > HAA
33 > imino acids 11 > BCAA 3 > Tau 1.0
residue 100-1 AA residues, L/A 2.30, M/G
0.15
(Jemil, Nasri,
et al. 2017)
Sardinelle (S. aurita) fresh
muscle
5% sh diet (w/w), 7-wks
serum TC ≤ 22%, serum TG ≤ 46%, AI ≤ 47%,
LDL-c 41%, hepatic damage (AST ≤ 41%, ALT ≤
30%, PAL ≤ 21%, LDH ≤ 44%),
HDL-c 29% in hypercholesterolemic diet fed rats
(Khaled et al.
2012)
Shark (S. mitsukurii and C.
plagiosum) fresh
healthy liver
50 µg S8300 peptide mL-1
degree of injury in STZinduced mouse insulinoma
βcells (NIT1β),
8,201 and 8,300 Da, NH2terminal sequence
(NH2 MLVGPIGAAKVVYEQ)
(F. J. Huang &
Wu, 2010)
3 − 10 mg S8300 kg-1 BW d-1, 4-wks (intraperitoneal)
hepatic (≤ 24%) and renal (≤ 35%) SOD,
body weight loss, blood glucose 28 49% and
HbA1c levels, plasma lipid levels (cholesterol ≤ 24%,
TG ≤ 61% and FFA ≤ 67%), hepatic (≤ 23%) and
renal (≤ 29%) MDA, FAS and FAS/GAPDH mRNA
expression in pancreas, FAS protein expression,
apoptosis of pancreatic tissue (degree of βcells
injury) dose dependently in alloxaninduced diabetic
rats
Skate (R. kenojei) skin,
commercial collagen
perperation
200 − 300 mg kg-1 BW d-1, 8-wks (oral)
liver, visceral and subcutaneous adipose tissue
weight (but not epididymis adipose tissue), lipid
droplet size, plasma TG (≤30%), NEFA (≤ 31%), and
LDL-c (≤ 42%), hepatic TG (≤ 25%), liver lipid
accumulation, hepatic expression of SREBP-1 (≤
18%), FAS (≤ 29%), ACC (≤ 39%), SREBP-2 (≤ 13%),
HMGCR (≤ 32%), and CYP7A1 (≤ 176%), leptin (≤
23%),
plasma HDL-c (≤ 320%), hepatic expression of
PPAR-α (≤ 159%), CPT-1 (≤ 163%), and p-AMPK
(156%), adiponectin (≤ 131%) in HFD fed obese rats
1,050 Da, CAA 31% > Gly 22% > imino AA
16% > BCAA 9%, L/A 0.45, M/G 0.01
(Woo et al. 2018)
Skate skin 200 mg kg-1 BW d-1, 8-wks (oral)
plasma and hepatic TG and TC, hepatic expression of
SREBP-1, FAS, ACC, and SREBP-2,
hepatic expression of MAPK, PPAR-α, ACOX1, and
CYP7A1 in obese rats
(H. Lee et al.
2018)
Smooth hound viscera food intake (except for undigested and hydrolysate),
plasma glucose levels (≤ 28%), uric acid (except <
1 kDa fraction) (≤ 27%), plasma TC (≤ 17%), TG (≤
23%), atherogenic index of plasma (AIP) (≤ 68%),
plasma HDL-c (≤ 61%), creatinine (≤ 16%) in
high-salt (18% NaCl) and -fructose (10%) diet
(HSFD)-induced hypertensive rats
CAA 38 − 41 > HAA 29 − 40 > EAA
33 − 34 > BCAA 11 − 12 > imino AA 7 − 9 g
100 g-1 AA, L/A 1.36 1.54, M/G
0.13 − 0.19
(Abdelhedi et al.
2019)
Thornback ray (R. clavata)
fresh muscle
700 mg kg-1 BW d-1, 1-mo
hepatic damage (AST ≤ 21%, ALT ≤ 27% and ALP ≤
15%), renal dysfunction (urea ≤ 20%, creatinine ≤
37%, uric acid ≤ 16%), plasma lipid prole (TG ≤
35%, TC ≤ 26%, LDL-c 49%, VLDL-c 45%), AI ≤
61%, blood glucose ≤ 25%, α–amylase ≤ 22%,
HDL-c 30% in hypercholesterolemic diet fed rats
(Lassoued et al.
2018)
Tuna extract 200 − 400 mg kg-1 BW d-1, 10-wks
BW, epididymal and abdominal adipose tissue
weight, liver weight, serum glucose (≤ 24%), TG (≤
28%), TC (≤ 23%), LDL-c (≤ 24%), insulin (≤ 28%),
leptin (≤ 35%), ALT and AST, hepatic expression of
lipogenesis and adipogenesis related genes (C/
EBP-α/-β/-δ, PPAR-γ, CD36, SREBP-1, LPL and FAS),
expression of GLUT4,
serum HDL-c (≤27%), hepatic expression of AMPKα,
and AMPKβ in HFD-fed obese rats
(Y. Kim etal.
2016)
Table 4. (Continued).
Fish name and part used Observed eect(s)
Characteristics of material and or peptide
(AA, sequence, and MW) Reference
(Continued)
20 S. ABACHI ETAL.
Warm sea sh skin,
commercial type I and
III collagen preparation
(Naticol®) by Weishardt,
France
4 g kg-1 BW d-1 20-wks
BW gain (fat- but not lean-mass), basal plasma
glucose, plasma cholesterol, plasma insulin
(non-signicant),
plasma TG in HFD fed obese rats
2 kDa, CAA 30% > Gly 30% > imino AA 23%
> BCAA 6%
L/A 0.39, M/G 0.04
(Astre et al. 2018)
Wild marine fresh sh
scales
1 g kg-1 BW every other day, 6-wks (intragastrical)
weight gain (8%), food intake, blood fasting glucose
(29%), relative level of insulin (34%) and GHbA1c
(36%) in obese hypercaloric diet fed rats
(Raksha et al.
2018)
Wild-caught chum salmon
skin
9 g kg−1 BW d-1, 28-d (intragastrical)
weight loss, leptin and resitin,
blood lipid and glucose metabolism, insulin
resistance, adiponectin levels, GLUT4 expression in
skeletal muscle, liver steatosis, PPAR-α expression in
live in hypercaloric (high-cholesterol/fat) diet
induced diabetic rats
130 − 3,000 Da (> 95%), GLPGPLGPAGPK (C. Zhu, Zhang,
Mu, et al.
2017)
Wild-caught chum salmon
skin
9 g kg-1 BW d-1, 4-wks
FBG, endothelial thinning and inammatory
exudation in carotid-artery vascular endothelial cells
in high-cholesterol, high-fat diet (31% beef tallow +
0.2% choline bitartrate)-induced diabetic rats
(C. Zhu, Zhang,
Liu, et al.
2017)
Wild-caught chum salmon
skin, collagenic
preparation
1.35 g kg−1 BW, 8-wks (intragastrical)
fasting insulin and glucose, TC and TG, serum
malondialdehyde, ultrastructural impairment of islet
in high fat diet induced hyperinsulinemic rats
(J. Wang et al.
2010)
Zebra blenny (S. basilisca)
fresh frozen muscle
400 mg kg-1 BW, 8-wks (gastric gavage)
serum and hepatic TG (≤ 38%), TC (≤ 70%), and
LDL-c (≤ 81%), hepatic damage (ALAT and ALP) (≤
38%), kidney damage (urea (≤ 24%), creatinine (≤
8%))
serum and hepatic HDL-c (≤ 54%) in
hypercholesterolemic diet fed rats
Lysates of crude proteases of zebra blenny
(3 − 7 kDa (34%), >10 kDa (29%)), smooth
hound (>10 kDa (54%), 3 − 7 kDa (12%),
and sardinelle (>10 kDa (41%), 7 − 10 kDa
(26%))
(Ktari et al. 2015)
Zebra blenny (S. basilisca)
muscle
400 mg kg-1 BW d-1, 21-d (gastric gavage)
serum and intestine α-amylase activity, blood
glucose ≤ 60%, HbA1c, serum lipid prole (TC ≤
38%, TG ≤ 62%, LDL-c 51%), hepatic lipid prole
(TC ≤ 65%, TG ≤ 38%, LDL-c 83%), myocardial
enzymes (CPK ≤ 32.9%, AST ≤ 44.6%, LDH ≤
32.7%), serum enzymes (ALT ≤ 46%, ALP ≤ 44%,
GGT ≤ 45%), serum bilirubin (T-Bili ≤ 46%, D-Bili ≤
63%), hyperglycemic hepatotoxicity, apoptosis of
pancreatic β-cells,
serum and hepatic HDL-c (23 38%) in
alloxan-induced diabetic rats
CAA 35% > BCAA 31% > sulfur containing
AA 3%, L/A 0.51
(Ktari et al. 2013)
Increased, improved, stimulated and or upregulated expressions are denoted as , and decreased, inhibited and or downregulated expression denoted as ”.
HAA: Hydrophobic amino acids (glycine, alanine, valine, isoleucine, leucine, phenylalanine, proline, methionine, and tryptophan), AAA: Aromatic amino acids
(phenylalanine, tryptophan, and tyrosine), PCAA: Positively charged amino acids (arginine, histidine, lysine); NCAA: Negatively charged amino acids (aspartic
acid, glutamic acid), EEA: Essential amino acids (phenylalanine, valine, threonine, isoleucine, methionine, histidine, leucine, lysine and tryptophan), BCAA:
Branched chain amino acids (isoleucine, leucine and valine), LDH: Lactate dehydrogenase, AOPP: Advanced oxidation protein product, ACACA: Acetyl-CoA
carboxylase A, SCD1: Stearoyl-CoA desaturase 1, ALT: Alanine aminotransferase, ALP: Alkaline phosphatase, GGT: Gamma-glutamyl transpeptidase, PON2:
Protein paraoxonase 2, VCAM1: Vascular cell adhesion protein 1, p-AMPK: Phosphorylated 5’ AMP-activated protein kinase, SGR: Specic growth rate, FER:
Feed eciency ratio, PER: Protein eciency ratio, PR: Protein retention, VSI: Viscerosomatic index, ACOT1: Acyl-CoA thioesterase, MUFA: Monounsaturated
fatty acids, FADS1: Fatty acid desaturase 1, sCT: Salmon calcitonin.
Table 4. (Continued).
Fish name and part used Observed eect(s)
Characteristics of material and or peptide
(AA, sequence, and MW) Reference
bromelain, pepsin, trypsin and pancreatin in producing
DPP-IV inhibitor peptides from Pacific hake, halibut, tilapia,
milkfish and salmon skin, irrespective of degree of hydrolysis
and process efficiency in terms of enzyme/substrate (E/S)
ratio (Li-Chan etal. 2012; T.-Y. Wang etal. 2015). Accordingly,
potent antidiabetic and anti-obesity biopeptides were pro-
duced from fish by the proteolytic activity of various Bacillus
species (Jemil, Nasri, et al. 2017; Nasri et al. 2018; Nasri
etal. 2015). Plasma glucose and hepatic glycogen levels were
rather more affected by B. mojavensis A21 digested goby fish
fillet protein than gray triggerfish digestive crude proteases
digested and non-digested proteins (Nasri et al. 2015).
Hypoglycemic goby fish derived proteins rather contained
similar amounts of amino acids, yet their L/A and M/G
ratios differed to significant extent (non-digested: 0.49, 0.13,
gray triggerfish digestive crude proteases digested lysate: 0.46,
0.15, B. mojavensis A21 digested lysate: 0.58, 0.19 respec-
tively) (Nasri etal. 2015). Corolase followed by alcalase and
combination of alcalase + flavorzyme were the most appro-
priate enzymes for production of hypoglycemic peptides from
salmon trimmings (Neves et al. 2017). In line with Neves
et al. study, among a wide variety of enzymes, pepsin,
CorolaseN or papain digested steelhead skin gelatinous prepa-
ration exhibited markedly strong anti-ACE and anti-DPP-IV
activity over others (Cheung and Li-Chan 2017; Neves etal.
2017). Nonetheless, gray triggerfish digestive crude proteases
digested peptides of goby fish were more potent antioxidants
enhancing superoxide dismutase (SOD), catalase (CAT), and
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 21
Table 5. Fish peptides with bioactivities on MetS associated risk factors (preclinical and clinical studies).
Fish/product name and
or protein source Experimental design
Subjects and inclusion
criteria Observation
Characteristics of
material and or
peptide (AA,
sequence, and MW)
Study parameter
(reference)
Atlantic cod (Gadus
morhua),
commercial preparation
by Firmenich Bjørge
Biomarin AS, Norway
20 mg sh kg-1 BW or
casein control, before
standardized breakfast
meal, double-blind
cross-over trial, 2-study
days with 4 7 d
wash-out in between
41 healthy and active
subjects, aged
41 − 64 y, BMI
20 − 30 kg/m2, no
food allergies or
intolerance to sh,
no anti-diabetes,
hypertension, chronic
diseases, and acute
infection medications
postprandial insulin
concentration
compared with casein
in subjects
<1 kDa (66%), HAA
348 > CAA
335 > BCAA
169 > Tau 7 mg g-1
protein, L/A 1.40,
M/G 0.43
(Dale et al. 2018)
Atlantic cod (Gadus
morhua),
commercial preparation
by Firmenich Bjørge
Biomarin AS, Norway
40 mg sh kg-1 BW d-1,
double-blind cross-over
trial, 1 week with
1-week washout periods
in between
31 healthy subjects,
aged 60 − 80 y, BMI
20 − 30 kg/m2, no
food allergies or
intolerance to sh,
no anti-diabetes,
hypertension, chronic
diseases, and acute
infection medications
serum glucose and
insulin levels in
subjects
CAA 335 > HAA
307 > BCAA
127 > Tau 7 mg g-1
of protein, L/A
1.40, M/G 0.43
(C. Jensen et al.
2019)
Atlantic herring (Clupea
harengus) head, gut,
backbone byproducts,
Atlantic salmon,
(Salmo salar)
backbone byproduct,
Atlantic cod (Gadus
morhua) muscle,
commercial tablet
preparation by Faun
Pharma AS, Vestby,
Norway
2.5 g protein d-1 8-wk
double-blind, randomized,
intervention study with
parallel group design
and four intervention
arms
93 overweight/ obese
adult subjects, aged
18 − 69 y, BMI >27 kg/
m2, FBG <
7.0 mmol/L, no
anti-inammatory or
glucose/lipid
metabolism
inuencing
medications
fasting and
postprandial serum
glucose, fructosamine/
albumin ratio (except
herring and salmon),
serum
α-hydroxybutyrate,
acetoacetate, total n-3
PUFA and n-3/n-6
PUFA ratio (except
salmon and cod),
serum
β-hydroxybutyrate
(except salmon)
fasting adiponectin
(except herring and
salmon), total SFA
(except cod)
compared to casein/
whey in overweight
subjects
Cod: CAA 1156 > EAA
1106 > HAA
1051 > BCAA
473 > imino acids
110 mg d-1, L/A
1.56, M/G 0.65,
Salmon: <4 kDa
(93%),
0.2 − 0.5 kDa
(14%),
HAA 1096 > C AA
928 > EAA
818 > BCAA
332 > imino acids
226 mg d-1, L/A
1.12, M/G 0.23,
Herring <4 kDa
(97%),
0.2 − 0.5 kDa
(14%), CAA
982 > EAA
782 > HAA
955 > BCAA
311 > imino acids
178 mg d-1, L/A
0.66, M/G 0.24
(Hovland et al.
2019)
Blue whiting
(M. poutassou), fresh
frozen muscle,
commercial food
supplement
preparation
containing Slimpro®
by Compagnie des
Pêches Saint Malo
Santé, France
one- or two-dose (1.4 g
Slimpro® dose-1) before
main meal (if one-dose),
before lunch and dinner
(if two-dose) sh and or
placebo (Slimpro®
replacement by whey
protein isolate)
treatment, monocentric,
randomized study, 90-d
120 (25% males, 75%
females) slightly
overweight Caucasian
subjects, aged
18 − 55 y, BMI:
25 − 30 kg/m2, mild
hypocaloric
(-300 kcal/day)
weight, fat mass, BMI,
extracellular water,
waist, hip, and thighs
circumference,
serum GLP-1 and CCK
levels in slightly
overweight subjects
Body weight control
(Nobile et al.
2016)
Cod protein, commercial
tablet preparation by
Science in Nutrition
Ltd., Bergen, Norway
6 cod or placebo tablets
d-1 (500 mg of cod
protein + 350 mg
stabilization and lling
agents (maltodextrin,
microcrystalline
cellulose, ascorbic acid,
and α-tocopherol) and
sweeteners (fructose
and vanilla) for 1st 4-wk
and 12 tablets d-1 for
2nd
4-wk, double-blind,
randomized, controlled
intervention study, 8-wk
40 subjects (20 males,
20 females (20 for
each intervention)
aged 20 − 70 y, BMI
27 kg/m2, FBG
<7 mmol/l, no
allergies conicting
with sh intake, no
medications aecting
blood pressure,
blood lipids or blood
sugar
2-h insulin C-peptide
level relative to
baseline, fasting and
2-h glucose level (at
end of study), serum
LDL-c
GHbA1c, palsma
HDL:LDL ratio
compared to placebo in
overweight adults
CAA 414 > BCAA
171 > imino AA
34.5 > Tau 7 g kg
sh protein,
L/A 1.47, M/G 0.50
(Vikøren et al.
2013)
(Continued)
22 S. ABACHI ETAL.
Fatty- (salmon, rainbow
trout, Baltic herring,
whitesh, vendace, or
tuna) or lean- (pike,
pike-perch, perch,
saithe, or cod) sh
≥ 4 sh portin wk-1 or
control (lean beef, pork,
or poultry and < 1 sh
portion wk-1) diet in
parallel with routine
clinic drugs as part of
regular coronary heart
disease treatment
including statins and
betablockers, controlled
randomized dietary
intervention study, 8-wk
35 coronary heart
disease subjects
(myocardial infarction
or unstable ischemic
attack diagnosed
patients in past ≤
3 y), aged 55 − 67 y,
BMI 24 − 30 kg/m2
size of HDL particles,
serum cholesterol,
cholesterol esters, TL
in very large HDL
particles, serum
n-3 FA, DHA, and ratio
of n-3 FA/ total FA
(except lean sh),
serum cholesterol,
cholesterol esters, TL
in very large HDL
particles (except fatty
sh) in coronary heart
disease subjects
Benecial eect of
sh intake on
lipoprotein
particles in
patients with
coronary heart
disease (Erkkilä
et al. 2014)
Fish (lean-sh cod,
pollock and haddock
skin), commercial
preparation of high
MW food/
pharmaceutical-grade
sh gelatin by
Norland Products,
United States
25% of total daily protein
from sh gelatin + n-3
PUFA and or n-3 PUFA
alone, crossover
randomized study,
2 × 8-wk experimental
periods, a 4-wk run-in
period and a 12-wk
washout period
21 subjects (10 males,
11 females), aged
35 − 70 y, 75 g oral
glucose tolerance
test, BMI > 25 kg/m2,
overweight/ obese,
insulin-resistant with
high fasting plasma
insulin, impaired
fasting plasma
glucose
(5.6 − 6.9 mM), and or
with had impaired
glucose tolerance
following 2-h post
75 g oral glucose
tolerance test with
plasma glucose
corresponding to
7.8 − 11.0 mM, no
food allergies or
intolerance to sh,
no lipid and glucose
metabolism
interfering
medications, no
current diabetes
hyperlipidemia,
hypertension, hepatic
or metabolic diseases
protein intake, hsCRP
in females (20%)
(except n-3 PUFA),
hsCRP males (13%)
(except gelatin + n-3
PUFA)
carbohydrate intake,
hsCRP in males (40%)
(except n-3 PUFA),
hsCRP in females (6%)
(except gelatin + n-3
PUFA), plasma TG in
females 8 − 23%
(gelatin + n-3 PUFA
more eective),
plasma TG in males
11 − 25% (n-3 PUFA
more eective)
in free-living
insulin-resistant
subjects
Reducing CVD risk
by lipid lowering
and
anti-inammatory
eects of sh
gelatin and n-3
PUFA
supplementation
in
insulin-resistant
subjects
(Picard-Deland
et al. 2012)
Fish and tuna Mean energy-adjusted
14.4 g d-1 sh intake,
cross-sectional study,
dish-based
semiquantitative food
frequency questionnaire
420 (female) subjects,
aged > 30 y
MetS (96%),
hypertriglyceridemia
(89%), low HDL-c
(43%) and blood
pressure (77%),
intake of total energy,
carbohydrates, and
rened grains in
female subjects
Alleviation of
metabolic
syndrome
(Zaribaf et al.
2014)
Lean seafood (cod,
pollock, saithe, and
scallops)
Lean-seafood (e.g., 150 g
pollock for lunch, 115 g
cod for dinner) or
no-seafood lunch and
dinner meals,
randomized controlled
trial with crossover
design, 2 × 4-wk
separated by a 5-wk
washout period with
3-wk run-in period
20 healthy Caucasian
subjects, aged
18 − 65 y, BMI
24.9 − 26.3 kg/m2
serum fasting and
postprandial TG
(fasting TG in
chylomicrons and
VLDLs, fasting VLDL
particle size,
postprandial
medium-sized VLDL
particles), TC/HDL-c
ratio in fasted and
postprandial serum,
fasting TG/
HDL-cholesterol ratio
(13%), fasting serum
transferrin receptors,
postprandial serum
C-peptide and lactate
concentrations in
subjects
Lean seafood: CAA
46 > HAA
41 > BCAA 18 > Tau
0.3%, L/A 1.25,
M/G 0.45
Benecial health
eects of
lean-seafood diet
on cardiovascular
lipid risk factors
and long-term
development of
insulin
resistance, T2D,
and CVD in
healthy subjects
(Eli Kristin Aadland
et al. 2015) (Eli
K Aadland et al.
2016)
Table 5. (Continued).
Fish/product name and
or protein source Experimental design
Subjects and inclusion
criteria Observation
Characteristics of
material and or
peptide (AA,
sequence, and MW)
Study parameter
(reference)
(Continued)
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 23
Salmon, Atlantic salmon,
sardine, trout, tuna
(skin on/ o,
marinated, avored,
canned, slices in
spring water, or
lightly seasoned
frozen sh llets)
4-servings wk-1 (800 mg
d-1 EPA + DHA), parallel
randomized controlled
study stratied by CRP
(< 3 and ≥ 3 mg/L) on
entry to study, 8-wk
80 healthy elderly
subjects, aged ≥
64 y, BMI ≥ 18.5 kg/
m2
total n-6 FA in plasma
PL,
very long chain (VLC)
n-3 PUFA in
erythrocytes, plasma
PL compared to red
meat in healthy
subjects
Benecial eect of
sh diet on
cardiovascular
biomarkers
(Grieger, Miller,
and Cobiac
2014)
Skate (Raja Kenojei)
skin, collagenous
preparation to
capsule form by
Serom Co., Ltd.
(Jeonnam, South
Korea)
2 g d-1 (4 × 0.5 g sh or
placebo capsule, twice
daily in morning and
evening), randomized,
placebo-controlled, and
double-blinded study,
12-wks
90 (17 males, 73
females) healthy
volunteer subjects,
aged 40 − 52 y, BMI
24 − 27 kg/m2
body fat (-1.2 kg, no
toxicity, and adverse
eects) in overweight
subjects
Treatment for
obesity and
related disorders
(reduction of
body fat) (Tak
et al. 2019)
Tuna (canned),
dark- and
light-eshed sh
0 – ≥ 6 sh serving d-1,
standardized and
validated
food-frequency
questionnaire, cohort
study with 26 y
follow-up
84,136 subjects (female)
aged 30 − 55 y, with
no known cancer,
diabetes mellitus,
angina, myocardial
infarction, stroke, or
other CVD
risk of coronary heart
disease by 1-serving
sh d-1 (24%) in
subjects
Fish and poultry
intake decrease
coronary heart
disease risk
(Bernstein et al.
2010)
Wild marine deep-sea
sh fresh meat,
Commercial
preparation (mixed
proteases)
6.5 g sh collagenous
protein or placebo
(carboxymethylcellulose)
d-1 before breakfast and
bedtime
in parallel with routine
clinic drugs (T2D:
anti-diabetic drugs
(tolbutamide
500 − 3000 mg/d and or
metformin
500 − 1500 mg/d),
hypertension:
anti-hypertensive drugs
(metoprolol
50 − 100 mg/d,
verapamil 80 − 240 mg/d,
or losartan
25 − 100 mg/d), no
insulin, insulin secretion
stimulators, anti-ACE
and anti-hyperlipidemic
medicines), 3-m,
randomized double
blind study
150 subjects (100 T2D
diabetic patients
with hypertension,
50 healthy
individuals (no
current T2D,
hypertension, T1D,
infectious diseases,
current and historical
CVD, liver, kidney,
cerebrovascular
diseases, cancer) for
each group), aged
64 − 68 y, BMI:
24 − 25 kg/m2, FBG
7.0 mmol/L, SBP ≥
140 mmHg, or DBP ≥
90 mmHg
FBG, HbA1c, fasting
insulin, creatinine,
serum TG, TC, LDL-c,
and FFA, Cyp450, PGI2
and NO,
insulin sensitivity
index, insulin
secretion, HDL-c,
bradykinin,
adiponectin compared
to patient control in
T2D and hypertension
subjects
130 − 3,000 Da (>
95%, mostly
300 − 800 Da)
Therapeutic eects
on glucose and
lipid metabolism,
insulin
sensitivity, renal
function in
Chinese patients
with T2D and
hypertension
(C.-F. Zhu, Li,
Peng, Li, etal.
2010)
Wild marine sh 6.5 g × 2 (before breakfast
and bedtime), 3-m,
blind randomized
(treatment and control
groups (50 per group))
concomitant with
antidiabetic medicines
(tolbutamide and or
metformin), and or
carboxymethylcellulose
placebo
100 T2D patients
(diagnosed when
fasting blood glucose
(FBG) concentration
was ≥ 7.0 mmol L−1)
and 50 healthy
subjects
FBG, GHbA1c, fasting
blood insulin, TG, TC,
LDL-c, FFA, hsCRP and
NO,
insulin sensitivity
index, HDL-c,
bradykinin, PGI2,
adiponectin in T2D
patients
95% of oligopeptides
MW 130 − 3,000 Da
(300 − 800 Da)
(C.-F. Zhu, Li, Peng,
Zhang, et al.
2010)
Table 5. (Continued).
Fish/product name and
or protein source Experimental design
Subjects and inclusion
criteria Observation
Characteristics of
material and or
peptide (AA,
sequence, and MW)
Study parameter
(reference)
(Continued)
24 S. ABACHI ETAL.
Wild marine fresh sh
Mixed proteases,
ultra-ltration,
desalination,
cryoconcentration
under vacuum at
70 °C, decolorization,
and lyophilization
6.5 g sh collagenous
protein or placebo
(water-soluble starch)
d-1 before breakfast and
bedtime in parallel with
routine clinic drugs
(T2D: anti-diabetic
drugs (tolbutamide
500 − 3000 mg/d and or
metformin
500 − 1500 mg/d),
hypertension:
anti-hypertensive drugs
(metoprolol
50 − 100 mg/d,
verapamil 80 − 240 mg/d,
or losartan
25 − 100 mg/d), no
insulin, insulin secretion
stimulators, anti-ACE
and anti-hyperlipidemic
medicines), 90-d,
random intervention
study
200 subjects (T2D
patients with/without
hypertension, 50
age-matched healthy
individuals (no
hypertension,
diabetes, coronary
heart disease, or
hyperlipemia) for
each group) subjects,
aged 62 − 68 y, FBG
7.0 mmol/L,
systolic blood
pressure (SBP) ≥
140 mmHg or
diastolic blood
pressure (DBP) ≥
90 mmHg,
FFA, cytochrome P450,
hsCRP, leptin, resistin,
adiponectin in T2D
with/without
hypertension subjects
130 − 3,000 Da (>
95%), 700 − 860 Da
(48%),
300 − 500 Da (28%)
(Cui-Feng etal.
2010)
Increased, improved, stimulated and or upregulated expressions are denoted as , and decreased, inhibited and or downregulated expression denoted as
. HAA: Hydrophobic Amino Acids (glycine, alanine, valine, isoleucine, leucine, phenylalanine, proline, methionine, and tryptophan), AAA: Aromatic Amino
Acids (phenylalanine, tryptophan, and tyrosine), PCAA: Positively Charged Amino Acids (arginine, histidine, lysine); NCAA: Negatively Charged Amino Acids
(aspartic acid, glutamic acid), EEA: Essential Amino Acids (phenylalanine, valine, threonine, isoleucine, methionine, histidine, leucine, lysine and tryptophan),
BCAA: Branched Chain Amino Acids (isoleucine, leucine and valine).
Table 5. (Continued).
Fish/product name and
or protein source Experimental design
Subjects and inclusion
criteria Observation
Characteristics of
material and or
peptide (AA,
sequence, and MW)
Study parameter
(reference)
glutathione peroxidase (GPX) and glutathione (GSH) activities
in liver and kidney tissues over B. mojavensis A21 digested
hydrolysate (Nasri et al. 2015). Overall, B. amyloliquefaciens
An6 fermented sardine flour was rather more hypoglycemic
and antioxidative than the B. subtilis A26 fermented muscle
proteins (Jemil, Nasri, et al. 2017).
The enzymatic hydrolysis enhanced anti-DPP-IV effect
of the non-hydrolyzed fish skin gelatins by about +5-fold
increasing it from about 10% to almost 45% − 50% (Li-Chan
et al. 2012; T.-Y. Wang et al. 2015). Similarly, enzymatic
lysis, alcalase, alcalase + flavorzyme, corolase except
PROMOD, increased the anti-DPP-IV activity of undigested
salmon proteins up to about +8-folds (Neves et al. 2017).
In agreement, enzymatic digestion, in particular with trig-
gerfish proteases, considerably enhanced the hypolipidemic
effects of undigested muscle proteins of goby fish (hepatic
TG and TC: up to 22%, serum TG: 18%, serum TC: 12%,
serum HDL cholesterol: 10%, serum LDL cholesterol: 56%,
serum VLDL cholesterol: 19%) (Nasri etal. 2018). The enzy-
matic digestion similarly enhanced the hypolipidemic and
hypoglycemic effect of thornback ray sarcoplasmic proteins
as high as 16% (TG: +16%, TC: +3%, LDL-c: +6%, VLDL-c:
+14%, HDL-c: +1%, aspartate transaminase (AST): +2%,
alanine transaminase (ALT): +14%, alkaline phosphatase
(ALP): +8%, blood glucose and α–amylase: +8%, AI: +5%)
(Lassoued et al. 2018). In contrary, enzymatic hydrolysis
lessened the metabolic benefiting effects of goby fish undi-
gested proteins, in terms of food intake, with L/A and M/G
ratios of 0.49 and 0.13 compared to the same ratios of the
triggerfish proteases digested (0.46 and 0.15) and B. mojaven-
sis A21 proteases hydrolyzed (0.58 and 0.19) fractions, by
−14% to −17% (Nasri et al. 2018). Evidently, triggerfish
crude proteases extract was a more appropriate choice in
comparison to B. mojavensis A21 proteases for isolation of
antihyperlipidemic peptides from goby fish (Nasri et al.
2018). The enzymatic hydrolysis moderately improved the
hypocholesterolemic effects of zebra blenny muscle tissue
(5% − 23%) (Ktari et al. 2015).
Glucoregulatory activity of sardine meat flour protein
was drastically improved by bacterial fermentation (Jemil,
Nasri, etal. 2017). This may be justified by lower branched
chain amino acids (BCAA), hydrophobic amino acids
(HAA), essential amino acids (EAA), L/A and M/G ratios,
and higher charged amino acids (CAA) and taurine content
of B. amyloliquefaciens An6 over B. subtilis A26 fermented
peptides (Jemil, Nasri, et al. 2017). Sardinelle and smooth
hound proteases digested fractions of zebra blenny, with
smaller degree of hydrolysis (DH), comparatively were more
potent over zebra blenny digested lysate in preventing the
hepatic damage as well as lowering the serum and hepatic
TG, TC, and LDL-c levels (Ktari etal. 2015). Nevertheless,
as observed in liver histopathological studies, hepatic damage
of zebra blenny protease hydrolyzed zebra blenny muscle
protein treated rats, but not the other lysates, was entirely
alleviated (Ktari etal. 2015). Papain comparatively enhanced
the hypocholesterolemic activity of Alaska pollock, compared
to its undigested protein, at mRNA expression level of cho-
lesterol 7 R-hydroxylase (AKA cytochrome P450 7A1)
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 25
(CYP7A1) in hypercaloric diet fed rats (Hosomi etal. 2012;
Hosomi et al. 2009).
Even though hydrolysis in many instances has improved
anti-MetS properties of fish biopeptides yet, there have
been some exceptions. Undigested proteins of bogue muscle
(L/A: 0.23, M/G: 0.26) commonly were more potent
anti-hyperlipidemic than its hydrolyzed counterpart (L/A:
0.33, M/G: 0.22) at the same concentration (500 mg kg−1
BW d−1) (Lassoued et al. 2014). Generally, selectively sep-
arated peptides, yet not always, present better anti-MetS
activity. Ultrafiltration (UF) fraction of <1,500 Da was most
potent over >2,500, and 1,500 2,500 fractions against
DPP-IV activity at the concentration of 1 mg mL−1 (T.-Y.
Wang et al. 2015). In an earlier study, small MW
salmon-extracted fraction (<1,000 Da) at 2 mg mL−1 simi-
larly inhibited more of DPP-IV activity over other fractions
with an inhibitory concentration 50% (IC50) value of
1.35 mg mL−1 (Li-Chan et al. 2012). The vasodilating
potency of siki head hydrolysate increased going through
different isolation and purification processes (from 4.1 to
0.01 mg of protein) by +410-times with the purification
factor of up to 79% (Martinez-Alvarez et al. 2007). Gel
filtration chromatography (GFC) slightly improved the
activity of hydrolysate by +3.8-folds (Martinez-Alvarez
etal. 2007). Nevertheless, non-fractionated extract of mack-
erel tended to be more potent hypolipidemic, up to 65%,
on plasma lipids (16% (TC), 41% (HDL-c), 65% (LDL-c))
than its fractions (Itou and Akahane 2009). Separation
techniques such as electro-separation also have been useful
in extracting potent hypoglycemic peptides with in-vitro
glucose uptake stimulatory effect from fish (Durand
et al. 2019).
Umamizyme sequentially digested spine proteins, rather
hyperlipidemic, with highest HAA, M/G and L/A ratios
(48%, 0.90 and 2.83, respectively) in comparison to other
two hydrolysates tended to increase serum and hepatic
lipids as well as liver’s fatty acid synthase (FAS) activity
and Δ9 desaturase gene expression (Vik et al. 2015). Acid
protease lysed spine proteins with moderate M/G and L/A
ratios compared to casein (0.69 vs. 1.57 and 2.12 vs. 2.97,
respectively) resulted in high hepatic polyunsaturated fatty
acids (PUFA) and ceramides levels, however, hepatic di-
and tri-acylglycerols concentrations were decreased (Vik
et al. 2015). Among the three enzymes utilized for diges-
tion of zebra blenny sarcoplasmic proteins, crude proteases
of zebra blenny viscera resulted in highest DH with lowest
L/A ratio of 0.43, and medium M/G ratio of 0.40, over
the other two (L/A and M/G ratios of crude proteases of
smooth hound and sardinelle were 0.53, 0.38, and 0.57,
0.42, respectively) (Ktari et al. 2013). Administration of
goby fish sarcoplasmic hydrolysates (B. mojavensis A21 and
gray triggerfish proteases), in particular gray triggerfish
proteases digested one with 23% DH and more smaller
size peptides, could impede weight gain better than the
daily dose of 20 mg fluvastatin per kg of body weight
(Nasri etal. 2018). L/A ratio of B. mojavensis A21 digested
hydrolysate is +21% (0.58) higher than the undigested
protein (0.48) (Nasri etal. 2015). Interestingly, while the
same hydrolysate lowered more of the plasma glucose level
in hypercaloric diet fed rats, the undigested material
increased it by +16% (Nasri et al. 2015). Almost same
trend was observed on the effectivity of hydrolysate and
undigested protein on α-amylase activity. While α-amylase
activity was significantly inhibited by hydrolysates,
Table 6. Fish peptides with bioactivities on MetS associated risk factors formulated into commercial products.
Trade name of product
Fish type and
preparation Claimed eect Manufacturer, country
Category and or
dosage form Reference
Amizate®, amino acid
(25-AA, > 120
peptides e.g., IP/LP,
KP, anserine)
complex
Fresh Norwegian
salmon, hydrolysate
(endogenous
proteases)
Benecial health
eects on diabetes
and cardiovascular
diseases
Zymtech Production
AS, Norway
Powder (Nesse etal. 2014)
Fortidium Liquamen® White sh (Molva
molva) autolysate
Lowering glycemic
index, antioxidant,
and anti-stress
Biothalassol, France Capsule (Guérard et al. 2010)
Naticol®, type-I and
type-III collagen
Warm sea sh skin,
enzymatic
hydrolysate
Obesity associated
disorders
Weishardt
International, France
Powder (Astre etal. 2018)
Nutripeptin™ Cod fresh/ frozen llet,
enzymatic
hydrolysate
Lowering glycemic
index, improving
weight loss, and
maintaining
cardiovascular
health
Copalis, France Powder (Mora and Hayes 2015)
SeaSource™, collagen Wild-caught deep-sea
sh, cod,
hydrolysate
Benecial on CVD risk
prole and bone
health
Norland Products
Incorporated,
United States
Powder (Picard-Deland etal.
2012)
Slimpro® Blue whiting
(M. poutassou), fresh
frozen muscle,
enzymatic
hydrolysate
Weight management
( serum GLP-1 and
CCK levels)
Compagnie des Pêches
Saint Malo Santé
(France)
Commercial food
supplement
(Nobile et al. 2016)
Wellnex®,
food grade type-D
collagen
Tilapia (Oreochromis
sp.) scale,
enzymatic
hydrolysate
Diabetes treatment Nitta Gelatin Inc.,
Japan
Powder (Iba et al. 2016)
26 S. ABACHI ETAL.
undigested material increased its activity (Nasri et al.
2015). Source of fish-collagen and preparation techniques
seem to be important on particular parameters of
anti-obesogenic proteins (E. J. Lee et al. 2017; Woo etal.
2018). Tuna skin byproduct collagenous material, prepared
by subcritical water, decreased epididymal adipocytes’ size
while commercial collagenous skate skin perpetration had
negligible effect on epididymis adipose tissue weight in
high fat diet fed obese rats and effects were tissue depen-
dent (E. J. Lee etal. 2017; Woo etal. 2018).
Treatment and prevention of MetS with sh and
sh biopeptides
As one of the main risk factors of MetS, dyslipidemia could
have primary (genetical mutations) or secondary
(lifestyle-related and other factors) causes (Chan, Barrett,
and Watts 2004). Beside sedentary lifestyle as the main
cause, other common ones could be diabetes mellitus, alco-
hol abuse, chronic kidney disease, hypothyroidism, primary
biliary cirrhosis and other cholestatic liver diseases, cigarette
smoking, anabolic steroids, human immunodeficiency virus
infection, and nephrotic syndrome (Chan, Barrett, and Watts
2004). Lipid abnormality and elevated plasma triglyceride
(TG) levels may be genetically acquired, primary hypertri-
glyceridemia, or be of secondary type in which high-fat
diet, obesity, diabetes, hypothyroidism, and some medica-
tions trigger the condition. Obesity may be the most com-
mon stimulant of dyslipidemia yet, type 2 diabetes (T2D)
and high alcohol intake are too frequent. Dyslipidemia, one
of the most significant and common risk factors of cardio-
vascular diseases (CVD) that characterizes insulin resistance,
is an important hallmark of MetS and in the long run
advances into T2D. Abnormal levels of atherogenic lipid
and lipoprotein would be diabetic dyslipidemia with a char-
acteristic overproduction of large very low-density lipopro-
tein (VLDL) particles that would recruit a series of changes
in lipoprotein levels and develop into T2D within several
years. Over time cholesterol, predominantly low-density
lipoprotein (LDL), buildup initiates the oxidization of vas-
cular endothelium’s particles triggering the establishment of
tumor necrosis factor-alpha (TNF-α) producing atheroscle-
rotic plaques that adhere to the arterial wall promoting
arteriosclerosis originating inflammation (Kawanishi et al.
2013). In response to inflammatory mediators, TNF-α, endo-
thelium expresses cell adhesion molecules (intercellular
adhesion molecule 1 (ICAM-1), vascular cell adhesion pro-
tein 1 (VCAM-1), and endothelial-leukocyte adhesion mol-
ecule 1 (ELAM-1)) through translocation of nuclear factor
kappa-light-chain-enhancer of activated B cells (NF-kB) (K.-
W. Choi et al. 2010). For successful management of dyslip-
idemia, visceral fat mass and hepatic lipogenesis must be
reduced while improving the insulin sensitivity. This treat-
ment can be approached by reducing hepatic secretion
improving catabolism of VLDL, improving hepatic secretion
reducing catabolism of high-density lipoprotein (HDL) and
or speeding the clearance of LDL up (Chan, Barrett, and
Watts 2004). Prescription of specific therapeutic route for
the treatment of hypertriglyceridemia has not generally been
easy due to occurrence of cardiovascular disease risk factors
e.g., low HDL-c, MetS, obesity, T2D, proinflammatory and
prothrombotic biomarkers along with the condition. In addi-
tion to CVD, more severe cases of dyslipidemia could poten-
tially increase the risk of acute pancreatitis therefore the
condition should be mediated by therapeutic lifestyle changes
followed by medication in a timely manner (Shemesh and
Zafrir 2019). In both MetS and T2D, dysfunctional lipopro-
tein lipase activity, elevated cholesteryl ester transfer protein
activity, plasma VLDL (with or without chylomicronemia)
and hepatic free fatty acid (FFA) flux all play significant in
the development of hypertriglyceridemia. It is noteworthy
that abdominal adiposity, a feature of ectopic fat syndrome
and obesity, is strongly associated with low grade chronic
inflammation along with abnormal hormone secretion and
several metabolic disorders. Likewise, hypercholesterolemia
could be diet-induced or a genetically acquired condition
(familial or pure hypercholesterolemia). High plasma LDL
cholesterol (LDL-c) concentration in familial hypercholes-
terolemia increases the risk of premature coronary heart
disease (CHD). Mono- or combination-therapies in conjunc-
tion with dietary adjustments could alleviate dyslipidemia.
Fibrates, statins, niacin, cholesterol-absorption inhibitors and
handful emerging medications are regularly practiced for
the treatment of hypertriglyceridemia (Yuan, Al-Shali, and
Hegele 2007). Similar drugs (hydroxy-3
-methylglutaryl-coenzyme A (HMG-CoA) reductase inhib-
itors (statins) along with niacin, bile acid sequestrants,
fibrates, and omega-3 fatty acids) and mechanical means of
LDL-c removal are common therapies for hypercholesterol-
emia (Raal and Santos 2012).
Long term insulin resistance would lead to the develop-
ment of hyperglycemia and hyperinsulinemia, the two main
predictors of T2D. Moreover, they would indicate future
CVD and metabolic impairments (Paneni, Costantino, and
Cosentino 2014). Endothelial dysfunction in addition to
the lipid triad of dyslipidemia (emergence of VLDL-c, high
plasma TG and low HDL-c) induced by irregular insulin
signaling contributes to plaque formation and build up
within arteries (Paneni, Costantino, and Cosentino 2014).
Obesity, dyslipidemia, and insulin resistance comorbidities
are frequently associated with CVD (Paneni, Costantino,
and Cosentino 2014). Non-adipose tissue lipid accumulation
in muscle and the liver takes part in pathogenesis of insulin
resistance and recent studies have confirmed the role of
muscle-specific insulin resistance in promotion of hepatic
lipogenesis, nonalcoholic fatty liver disease, and atherogenic
dyslipidemia (Jornayvaz, Samuel, and Shulman 2010).
Persistent insulin resistance leads to impaired glucose and
lipid metabolism inducing oxidative stress followed by sub-
sequent immune response and cell damage. Impairment of
insulin signaling, and hyperglycemia are stressors of early
atherosclerotic lesion formation (Paneni, Costantino, and
Cosentino 2014). These lesions in diabetics, compared with
similar lesions of nondiabetics, advance into the ones with
large necrotic cores and higher macrophage content which
rather are correlated with glycated hemoglobin A1c
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 27
(GHbA1c) than lipid concentrations (Burke et al. 2004).
According to a community-based study, glycated hemoglo-
bin (≥ 6.0%) is greatly associated with the risk of CVD
than is with diabetes hence proven to be a better diagnostic
measure over fasting glucose for the evaluation of long-term
risk of successive CVD (Selvin et al. 2010). T2D enforces
significant economic burden on societies worldwide. T2D
related complications cost France €8.5 billion annually
(Emery et al. 2020). In the year 2003, direct medical costs
of T2D in France were estimately about €19.5 billion
(Emery et al. 2020).
One of the most common ways for treatment of diabetes
is the inhibition of enzymes involved in the digestion of
carbohydrates, α-amylase, and α-glucosidase, and or dipep-
tidyl peptidase 4 (DPP-IV) with diverse active and inactive
binding sites (supplementary Table 2). Diabetes is also
treated with glucagon-like peptide 1 (GLP-1) analogues,
sulfonylureas, meglitanides, basal insulins, thiazolidinediones
and sodium/glucose cotransporter 2 (SGLT2) inhibitors as
mono- or combination-therapies. Anti-DPP-IV medications
for example can be prescribed in combination with met-
formin and or insulin to control T2D and reduce GHbA1c
levels (Emery et al. 2020).
Statins (HMG-CoA reductase inhibitors) are rather
well-tolerated medications and among the most common
lipid-lowering compounds, yet this class of drugs are asso-
ciated with skeletal muscle, metabolic, neurological, and
other viable adverse effects. These side effects limit their
use for medication of hypercholesterolemia and manage-
ment of CVD risks. Per survey, 60% of former consumers
of the drug reported statin-associated muscle symptoms
which eventually made it impossible to continue the statin
therapy. The noncompliant patients taking less than 80%
of the prescribed therapy were 15% more in the risk of
CVD events not to mention the 45% increase in all-cause
mortalities (Thompson et al. 2016). Furthermore, many
adverse drug and or disease interactions have frequently
been reported for glucose lowering agents (Triplitt 2006).
This class of drug is well known for its weight gain side
effect making the management of T2D in overweight and
obese individuals difficult (Scheen and Van Gaal 2014).
Alternatively, synthetic drugs could possibly be substituted
with safe and or less damaging natural remedies and bio-
actives. In the following sections, authors will review the
in-vitro, in-vivo and human studies investigating the
anti-diabetes, anti-hyperlipidemia, and anti-obesity effects
of fish biopeptides.
Anti-diabetes peptides
In the subsequent two sub-sections, the anti-diabetic effects
of fish and fish derived proteins on mainly the enzymes
involved in glucose metabolism, fasting and postprandial
glucose and insulin levels (e.g., DPP-IV, α-amylase, and
α-glucosidase), GHbA1c, GLP-1 secretion, glucose trans-
porter type 4 (GLUT4) expression, glucose uptake and
energy expenditure in various experimental settings (in-vitro,
cell, animal, and human studies) would be discussed.
In-vitro and in-vivo studies
More than 50% ( 37 out of 65) of the studies reviewed
herein on the antidiabetic effect of fish biopeptides utilized
byproducts of a fish type e.g., skin, scale, frames, process
juice, etc. as substrate and starting material in their in-vitro
and in-vivo investigations (Tables 3 and 4). Many fish species
were studied for their glucoregulatory activity, yet salmon
was the most frequently tested type followed by cod, sardine,
and tuna. According to numerous observations, salmon
hydrolysates could beneficially modulate the activity of
enzymes involved in glucose metabolism, fasting and post-
prandial glucose and insulin levels, GLP-1 secretion, glucose
uptake and energy expenditure, GLUT4 expression in several
in-vitro and in-vivo studies whether in different cell lines
and or experimental animal models (Chevrier et al. 2015;
Drotningsvik et al. 2016; Drotningsvik, Pampanin, et al.
2018; Harnedy et al. 2018a; Jin 2013; Li-Chan et al. 2012;
Medenieks and Vasiljevic 2008; Neves et al. 2017; Pilon
et al. 2011; Roblet et al. 2016; Slizyte et al. 2016; J. Wang
etal. 2010). Oral dose of circulating salmon calcitonin (sCT)
has shown glucoregulatory activity affecting glucose and
insulin levels attenuating hyperglycemia along with better
islet β-cells function in diabetic and obese rats (Feigh etal.
2012; Feigh et al. 2011). Correspondingly, a 20% salmon
protein intake increased the sCT levels in hypercaloric diet
fed rats which may partially be the reason for beneficial
effects of this fish (Pilon etal. 2011). Anti-obesogenic effects
of sCT by means of reducing food intake and increasing
energy expenditure thus controlling body weight are also
of note (Lutz etal. 2000). Insulinotropic activity of salmon
was via ATP-sensitive potassium (KATP ) channel-dependent
and protein kinase A (PKA) pathways hence boosting the
membrane depolarization and intracellular calcium and
cyclic adenosine monophosphate (cAMP) concentration in
BRIN-BD11 cells (Harnedy et al. 2018a).
Salmon skin gelatinous hydrolysate diet eased the dia-
betic symptoms in rats. Polyphagia of streptozotocin (STZ)-
induced diabetic rats was significantly reduced by about
20% compared with the control group throughout 5-weeks
experiment period (Hsieh et al. 2015). Nevertheless, water
intake during the analysis insignificantly differed between
the normal non-diabetic rats and the fish-protein fed dia-
betic animals (Hsieh et al. 2015). Salmon skin gelatinous
hydrolysate diet reduced postprandial blood glucose level
to that of non-diabetic normal group thus area under curve
of fish and or normal diet fed groups were insignificantly
different from one another (Hsieh et al. 2015). The same
salmon diet tended to increase the plasma active GLP-1
levels in diabetic rats compared to diabetic and non-diabetic
control groups without inducing the secretion of hormones
(Hsieh etal. 2015). In previous studies, DPP-IV inhibitors
(e.g., sitagliptin, ASP8497 vildagliptin, and valine pyrroli-
dide), comparable to gelatinous hydrolysate, have demon-
strated protective effect and prevented the degradation of
active GLP-1 by DPP-IV thus improving its circulating
plasma levels and exerting glycemic control in similar dia-
betic experimental animal models (Hsieh et al. 2015; S.-J.
Kim, Nian, etal. 2009; Larsen etal. 2003; Matsuyama-Yokono
28 S. ABACHI ETAL.
etal. 2009). Moreover, gelatinous salmon hydrolysate, sim-
ilar to a hypoglycemic DPP-IV inhibitor (ASP8497), could
reduce the non-fasting blood glucose levels effectively in
rats (Hsieh et al. 2015; Matsuyama-Yokono et al. 2009).
Percent DPP-IV activity in diabetic fish-gelatin diet fed rats
was reduced by 32.9% in comparison to the diabetic normal
diet fed rats and in another study comparable effect (50%
inhibition) was observed on equivalent experimental model
(STZ-induced diabetic rat) by long-term administration of
sitagliptin, an orally administered DPP-IV inhibitor (Hsieh
etal. 2015; S.-J. Kim, Nian, etal. 2009). Five-weeks admin-
istration of fish-gelatin hydrolysate diet may reverse the
pancreatic β-cell destructive hyperglycemic causing feature
of streptozotocin thus inducing insulin secretion in diabetic
mice comparable to that of commercial DPP-IV inhibitors
(ASP8497, vildagliptin, sitagliptin and isoleucine thiazoli-
dide) which have been reported for their positive qualitative
and quantitative effect on small pancreatic islets,
glucose-dependent insulin excretion and subsequent blood
glucose depressing effects (Hsieh et al. 2015; S.-J. Kim,
Nian, etal. 2009; Matsuyama-Yokono etal. 2009; Pospisilik
et al. 2003). Plasma insulin-to-glucagon ratio was 40%
lower in salmon skin hydrolysate diet fed diabetic rats (7)
compared with normal control group (12) and 4-times
higher compared with diabetic control (1.7) thus proving
beneficial on hypoinsulinemia (Hsieh etal. 2015).
Cod’s glucoregulatory effect was rather like salmon.
According to the observations, cod protein could, similar
to salmon, beneficially modulate fasting and postprandial
glucose and insulin levels, GLP-1 secretion, glucose clearance
and energy expenditure in several in-vitro and in-vivo studies
yet to the knowledge of authors its activity on carbohydrate
digesting enzymes and or DPP‐IV has not been tested
(Geraedts et al. 2011; I.-J. Jensen et al. 2016; Lavigne,
Marette, and Jacques 2000; Lavigne et al. 2001; Myrmel
etal. 2019; Tastesen etal. 2014; von Post-Skagegård, Vessby,
and Karlström 2006). Though cod protein did not affect
insulin-mediated glucose infusion rates (GIR60–120) in obese
rats, its effects were by some means tissue specific (Lavigne
etal. 2001). Cod protein increased insulin-stimulated glucose
uptake in white tibialis, white gastrocnemius, red gastroc-
nemius, quadriceps, extensor digitorum longus (EDL) mus-
cle, heart and as well slightly in brown adipose tissue (BAT)
(Lavigne etal. 2001). Contrarily, in the study of Drotningsvik
etal. according to biochemical analysis neither of the diets,
cod- nor casein-diet (20% protein from casein/whey (90%
casein, 10% whey), could demonstrate glucoregulatory effect
(glucose and insulin concentrations, glucose to insulin ratio,
serum α-amylase) in obese Zucker fa/fa rats (Drotningsvik
et al. 2015).
Other fish types exerted similar effects to salmon and
cod. All three zebra blenny hydrolysates were potent
α-amylase inhibitors, and their activities were only insig-
nificantly different from one another (zebra blenny crude
protease digested hydrolysate with lowest IC50 of 90 μg mL−1)
(Ktari etal. 2013). Blue whiting muscle proteins stimulated
the secretion of CCK secretion from STC-1 cells, but
non-hydrolyzed material tended to be ineffective (Cudennec
etal. 2012). Tilapia collagen peptide at 1.7 g kg−1 BW could
positively affect the diabetic rat better than metformin in
terms of diabetic symptoms and hyperglycemia (R. Zhang,
Chen, Chen, etal. 2016). Fish proteins like goby fish lysates
could also enhance renal function and attenuate hyperfil-
tration of uric acid treating hypercaloric-diet induced hyper-
glycemia in liver and kidney of experimental animals (Nasri
et al. 2015).
Clinical studies
Bioavailability and bioefficacy of glucoregulatory fish bio-
peptides, processed and unprocessed, were further tested in
several controlled human studies. Of all the fish types, cod
was tested the most for its health effects in healthy and ill
human subjects. Cod successfully exerted its health effects
on various diabetes factors (e.g., insulin sensitivity (M/I),
disposition index (β-cell function × (M/I)), fasting and post-
prandial glucose and insulin, GLP-1 and HbA1c) in numer-
ous sets of populations (Eli K Aadland et al. 2016; Eli
Kristin Aadland et al. 2015; Dale et al. 2018; Mortensen
et al. 2009; Ouellet et al. 2007; Ouellet et al. 2008; Ramel,
Parra, et al. 2009; Vikøren et al. 2013). Cod fillet diet
enhanced insulin sensitivity in overweight obese
insulin-resistant human subjects compared to lean beef,
pork, veal, eggs, milk, and milk products (BPVEM) (Ouellet
et al. 2007; Ouellet et al. 2008). Additionally, cod diet
exerted anti-inflammatory effects on high-sensitivity
C-reactive protein (hsCRP), an inflammation marker known
to be associated with T2D and CVD risk, in a similar pop-
ulation as previous study (Ouellet etal. 2007; Ouellet etal.
2008). Cod protein, as part of a composite meal (45 g pro-
tein), in a randomized meal study showed glucoregulatory
effects on glucose and insulin responses in healthy subjects
(von Post-Skagegård, Vessby, and Karlström 2006).
Throughout the double-blind, randomized, controlled inter-
vention study, ingestion of 3 g codfish tableted protein per
day did not affect fasting insulin, fasting insulin C-peptide
and homeostatic model assessment of insulin resistance
(HOMA-IR), however, 2-h postprandial glucose levels at the
time point of 8-weeks were substantially lower in fish con-
sumed overweight adults group compared to the placebo
group (Vikøren etal. 2013). Opposite to triggerfish hydro-
lysates which decreased GHbA1c level in serum and liver
alloxan-induced diabetic rats, upon 8-weeks consumption
of cod protein the hemoglobin’s level was significantly
increased (Siala etal. 2016; Vikøren etal. 2013). Additionally,
cod muscle proteins exhibited glucoregulatory and
anti-obesogenic activity tending to decrease fasting and post-
prandial serum glucose and fructosamine/albumin ratio,
increasing fasting adiponectin concentration compared to
casein/whey in overweight subjects (Hovland et al. 2019).
Single cod protein dose (6.6 mg g−1 of protein), compared
to casein, however, could just depress the postprandial insu-
lin secretion but wasn’t effective on blood glucose response
or GLP-1 levels in middle-aged healthy population (Dale
et al. 2018).
In the studies of Zhu etal., collagen and gelatin prepa-
rations from various fish presented hypoglycemic and anti-
diabetic effects in diabetic patients with and without primary
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 29
hypertension (C.-F. Zhu, Li, Peng, Li, etal. 2010; C.-F. Zhu,
Li, Peng, Zhang, etal. 2010). A commercial small molecular
weight fish collagenous hydrolysate preparation demonstrated
therapeutic effects on glucose and lipid metabolism, insulin
sensitivity as well as renal function in Chinese patients with
T2D and hypertension (C.-F. Zhu, Li, Peng, Li, etal. 2010).
Effects were similar in diabetes-only subjects where fasting
glucose and insulin as well as GHbA1c decreased in patients
with the intake of 13 g collagen per day (C.-F. Zhu, Li, Peng,
Zhang, et al. 2010). Moreover, comparable findings were
confirmed in another study in which similar fish collagenic
preparation beneficially modulated the molecules involved
in pathogenesis of diabetes and hypertension (Cui-Feng
et al. 2010).
Anti-hyperlipidemia peptides
In the succeeding two sub-sections, anti-hyperlipidemia
effects of fish and fish derived proteins, mainly on the
enzymes involved in lipid catabolism and metabolism,
hepatic expression of lipogenic genes, cholesterol homeosta-
sis associated hepatic genes, hepatic and serum lipid levels,
hepatic FA desaturase indices, BA excretion, AI and CIR,
hepatic and renal tissue damage prevention and protection,
in various experimental settings (in-vitro, cell, animal and
human studies) would be discussed.
In-vitro and in-vivo studies
Of all the studies focusing on hypolipidemic effects of fish
peptides about 40% (25 out of 62) made use of fish byprod-
ucts (Tables 3–5). Numerous fish were investigated for their
anti-hyperlipidemic activities; however, the most studied
type was Alaska pollock followed by sardine and salmon,
respectively. In-vivo analysis of Alaska pollock
anti-dyslipidemia peptides has shown to effectively control
lipid metabolism and levels in liver and plasma, cholesterol
homeostasis associated hepatic genes, hepatic FA desaturase
indices, fecal BA excretion and mRNA levels of Ileal Bile
Acid Transporter (IBAT) in pigs, rats, mice and or in fish
(Cai et al. 2015; Hosomi et al. 2010; Hosomi et al. 2011,
2012, 2013; Kato etal. 2011; Maeda etal. 2015; Shukla etal.
2006; Spielmann et al. 2009; Xu et al. 2016; Zheng et al.
2014). Sardine peptides similarly modulated the hepatic
enzymes involved in lipid metabolism, hepatic and plasma
lipid parameters, lipid vacuole formation, AI and CRI,
hepatic and renal tissue damage triggering enzymatic activ-
ities in various rat models (Affane et al. 2018; Athmani
et al. 2015; Benomar et al. 2015; Jemil, Abdelhedi, et al.
2017; Jemil, Nasri, et al. 2017; Khaled et al. 2012; Madani
et al. 2012). Hypolipidemic biopeptides of salmon alone or
in combination with fish oil as single or mixed fish protein
diet acted similar to Alaska pollock and sardine diets in
regulating the lipid parameters, lipid catabolism, lipid bio-
synthesis associated enzymes and their activity as well as
the hepatic expression of lipogenic genes in experimental
mice (Ait-Yahia et al. 2003; Drotningsvik et al. 2016; Saito
etal. 2009; Vik etal. 2015; Wergedahl etal. 2009; Wergedahl
et al. 2004; Dalila Ait Yahia et al. 2003; D Ait Yahia
et al. 2005).
Alaska pollock fillet protein not only modulated the
expression of hepatic genes involved in the lipid metabolism
but also affected serum and hepatic lipid levels in normal-fat
and high-fat hypercholesterolemic diet fed mice (supple-
mentary Table 3) (Hosomi et al. 2009). Alaska pollack
hydrolysate was more effective on liver lipid indices of
hypercholesterolemic diet fed rats (cholesterol: −27%, TG:
+6%) than normal diet fed ones (cholesterol: +2%, TG:
−12%) compared to casein, regardless of its significance
(Hosomi etal. 2012). Liver of hypercholesterolemic fed rats
had higher levels of cholesterol (+13-fold and +10-fold in
casein and fish diets, respectively) and TG (4-fold and 5-fold
in casein and fish diets, respectively) which may have been
the reason for higher organ weight (Hosomi et al. 2012).
Serum, liver, and fecal biochemical parameters as well as
liver FA (C16:0, C16:1n-7, C18:1n-9, C18:2n-6, C20:3n-6,
and C20:4n-6) and FA desaturase indices (C16:1/C16:0 and
C18:1/C18:0) were considerably modulated by different pro-
tein dietary sources (casein and Alaska pollock) (Maeda
et al. 2015). Nonetheless, growth parameters, organ weight,
hepatic fatty acid metabolism related activities of enzymes
and relative mRNA expression levels were not as much
affected by the purified protein diets, except for the signif-
icant downregulation of stearoyl-CoA desaturase 1 (SCD-1)
by fish proteins (positively correlated with hepatic C18:1/
C18:0 and C16:1/C16:0 ratios) in KK-Ay rats (Maeda etal.
2015). Findings of Maeda et al. are in comparative agree-
ment with Hosomi etal.s observations (Hosomi etal. 2013;
Hosomi et al. 2009; Maeda et al. 2015). Water-insoluble
proteins of Alaska pollock comparably exhibited hypocho-
lesterolemic effect compared to casein (M/G: 1.22, L/A: 2.7)
in cholesterol-free fed ovariectomized rats (Kato etal. 2011).
Smaller M/G ratio, compared to smaller L/A value, played
more vital for hypocholesterolemic effect of Alaska pollock
protein (L/A: 2.0, M/G: 0.38 (low-Met) and 0.76 (high-Met))
versus casein (L/A: 2.7, M/G: 1.22) (Kato et al. 2011).
Water-soluble proteins of some fish types such as blue whit-
ing similarly tend to demonstrate significant hypocholester-
olemic effect in obese animal models (Drotningsvik, Vikøren,
et al. 2018).
Inclusion of small amount of Alaska pollock could also
make significant changes in the hyperlipidemic state of
animal models. In the study of Hosomi et al., a diet of
10% Alaska pollock fillet protein significantly enhanced
the activity of carnitine palmitoyl transferase 2 (CPT-2),
however, its effect on acyl-CoA oxidase (ACOX), FAS and
glucose-6-phosphate dehydrogenase (G6PDH) were insig-
nificant compared to casein diet in mice models (Hosomi
etal. 2013). In accord, relative mRNA expression levels of
lipid metabolism related enzymes and transporters (i.
ATP-binding cassette transporter A1 (ABCA1), ii.
3-hydroxy-3-methylglutaryl coenzyme A reductase
(HMGCR), iii. ABC transporter G5 and G8 (ABCG5 and
ABCG8), iv. CYP7A1, v. LDL receptor (LDLR), and vi.
scavenger receptor B1 (SRB1)) were marginally affected by
30 S. ABACHI ETAL.
fish diet fed rats compared to casein, except SCD1 which
was downregulated by 8% (Hosomi etal. 2013). In addition
to hepatic cholesterol levels, relatively similar to the study
of Hosomi et al., relative mRNA concentrations of genes
involved in fatty acid metabolism in liver and adipose
tissue of fish protein fed pigs (acetyl-CoA carboxylase
(ACC) and FAS in liver and adipose tissue, peroxisome
proliferator-activated receptor alpha (PPARα) (and its target
genes ACOX and CPT-1a) in hepatic fatty acid oxidation),
except CPT‐1a, were insignificantly affected by raw Alaska
pollack fillet diet (Spielmann et al. 2009). Unlike undi-
gested protein, effect of papain digested protein of the
same fish was more prominent (+36%) on the relative
mRNA expression level of CYP7A1 than other cholesterol
metabolism associated genes (Src homology region 2
domain-containing phosphatase-1 (SHP-1), HMGR,
acetyl-CoA acetyltransferase (ACAT-1), LDLR, sterol reg-
ulatory element-binding protein 2 (SREBP-2), farnesoid X
receptor alpha (FXRα), liver receptor homolog-1 (LRH-1))
compared to casein in hypercholesterolemic diet fed rats
(Hosomi et al. 2012).
In addition to upregulation of antioxidation enzymes,
sardine muscle proteins at high and low doses in short-and
long-term controlled animal studies alleviated hyperlipidemia
in hypercaloric fed diet hyperglycemic and oxidative stressed
rats (Affane etal. 2018; Athmani etal. 2015; Benomar et al.
2015; Jemil, Abdelhedi, etal. 2017; Jemil, Nasri, etal. 2017;
Khaled etal. 2012; Madani etal. 2012). Interestingly, sardine
byproduct isolated proteins were more hypolipidemic than
its fillet proteins, by up to +73%, in terms of anti-obesity
effects, fecal lipid and cholesterol excretion (Affane et al.
2018). Likewise, effect of sardine byproduct was superior
(serum Paraoxonase-1 (PON-1): +119%, HDL2-PON-1:
+100%, HDL3-PON-1: +90%) to its fillet peptides conse-
quently affecting lipid peroxidation (serum thiobarbituric
acid reactive substances (TBARS): −34%, VLDL-TBARS:
−31%, LDL-HDL1-TBARS: −54%, HDL2-TBARS: −64%,
HDL3-TBARS: −63%) in obese rats (Affane et al. 2018). It
is noteworthy that lipid metabolism was more influenced
by bogue hydrolysate than sardine hydrolysate in hypercho-
lesterolemic diet fed rats (Benomar etal. 2015). Fish lysates
modulated the lipid profile of lipoproteins ((VLDL (TC: ≤
−80%, phosphatidylcholine: ≤ −60%, sphingomyelin: ≤
+248%), LDL (TC: ≤ −45%, phosphatidylcholine ≤ −50%,
sphingomyelin: ≤ +53%) and HDL (TC: ≤ −4%, phospha-
tidylcholine: ≤ −15%, sphingomyelin: ≤ +59%)) and serum
activity of HDL-associated enzymes (lecithin cholesterol
acyltransferase (LCAT) activity: +23% by bogue hydrolysate
and −35% by sardine hydrolysate) in hyperlipidemic animals
significantly yet bogue hydrolysate was more potent than
sardine (Benomar etal. 2015). Accordingly, bogue hydroly-
sate fed rats had 3.3-times higher fecal cholesterol excretion
rate than sardine (Benomar etal. 2015).
Salmon backbone hydrolysate tended to decrease lipo-
genesis involved enzymes increasing the hepatic PUFA levels
affecting the regulation of lipid and glucose metabolism
compared to casein diet in high fat fed animals (Vik et al.
2015). The same salmon off-cuts hydrolysate, at different
concentrations, in studies of Bjørndal et al. and Parolini
etal. downregulated the mRNA level of Scd1 in two different
animal models, chronically inflamed and apolipoprotein E
(ApoE)-deficient rats (Bjørndal et al. 2013; Parolini et al.
2014). Expression of acetyl-CoA carboxylase A (ACACA)
was considerably downregulated in 5% salmon spine hydro-
lysate fed ApoE-/- mice for 12-weeks while higher intake of
same diet (15%) for 2-weeks, did not affect its expression
in humanized (h)TNF-α rats (Bjørndal et al. 2013; Parolini
etal. 2014). Salmon diet increased plasma levels of carnitine
and its precursor (γ-butyrobetaine), short-chained and
medium-chained acylcarnitine esters and fatty acid compo-
sition in liver and white adipose tissue (WAT) (Bjørndal
etal. 2013). In particular, Δ5 desaturase index for n-3 fatty
acids and Δ6 desaturase index for n-3 fatty acids and n-3
index were higher in the earlier mentioned organ and tissue
of chronically inflamed rat (Bjørndal etal. 2013). A signif-
icant decrement in the lipid profile, namely TC, TG, and
LDL-c but not HDL-c, of hemodialysis patients upon car-
nitine supplementation of 750 mg per day was observed
(Naini et al. 2012).
Fatty acid metabolism of the obese animals was likewise
considerably affected by cod-diet compared to casein diet;
total n-3 PUFA level in epididymal WAT (eWAT), n-3/n-6
PUFA ratio in serum, liver and eWAT, and anti-inflammatory
fatty acid index (AIFAI) in serum, liver, skeletal muscle and
eWAT (Drotningsvik etal. 2015). Menhaden oil-cod protein
combined diet, however, could not reduce serum TG levels
compared with other protein-lipid diets in rats (Demonty,
Deshaies, and Jacques 1998). In agreement, cod diet (23%
(w/w) protein diet) had insignificant effect on fasting plasma
concentrations of non-esterified fatty acids (NEFA) and TG
in high fat diet fed rats (Lavigne etal. 2001). Nonetheless,
cod-diet was superior to casein-diet (14% and 36% higher
L/A and M/G, respectively) in reducing the serum concen-
tration of NEFA (Drotningsvik et al. 2015). In contrary,
neither cod- nor casein-diets (20% protein from casein/whey
(90% casein and 10% whey)) could have hypolipidemic
effects (insignificant effects on lipase and alanine transam-
inase concentrations, plasma TNF-α levels, serum TC, HDL/
LDL cholesterol, total BA and triacylglycerol concentrations)
in obese Zucker fa/fa rats (Drotningsvik etal. 2015). Plasma
HDL-c and LDL-c levels were heightened in cod/scallop fed
rats, as well as mRNA expression of sterol regulatory
element-binding transcription factor 1 (SREBF1) and
HMGCR, compared to low fat fed and other protein group,
however, other parameters of plasma metabolites and liver
lipids were not significantly affected (Tastesen et al. 2014).
Of all the monitored parameters only HDL: TC ratio was
non-negligibly higher in casein fed rats, than low-fat and
other protein diets (Tastesen et al. 2014). Evidently, bene-
ficial metabolic effect of 13-weeks cod/scallop diet consump-
tion was superior over chicken intake in alleviating aorta
atherosclerotic plaque burden in ApoE-/- rat (I.-J. Jensen
et al. 2016). Fatty seafood regimen (eicosapentaenoic acid
(EPA) and docosahexaenoic acid (DHA) rich) as well as
lean ones have repeatedly been proposed for their hypotri-
glyceridemic effects and subsequent lesser aorta
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 31
atherosclerosis (Liaset etal. 2019). Nevertheless, these effects
could not solitary be beneficial to the condition since diets
with comparable cholesterol content, but different amino
acid composition, have affected the plaque burden differently
(I.-J. Jensen etal. 2016; Tastesen et al. 2014). There seems
to be simply an association of diet type, in terms of protein
source, with metabolic benefiting effects important in pre-
vention and management of cardiometabolic syndrome.
Other fish types likewise demonstrated small and or large
hypolipidemic effects improving dyslipidemia, its associated
disorders and organ damages. Compared to Crestor
(cholesterol-lowering drug at the dosage of 10 mg kg−1 BW)
zebra blenny muscle hydrolysates more efficiently reversed
kidney (urea 19%, creatinine 2%) and hepatic damage (ala-
nine aminotransferase (ALAT) 22%, ALP 23%) while posi-
tively modulating the serum and hepatic lipids (serum: TG
+41%, TC +68%, LDL-c + 148%, HDL-c + 19%, and liver: TC
+37%, LDL-c + 23%, and HDL-c + 135%) in hyperlipidemic
animals (Ktari et al. 2015). Evidently, hepatic damage, in
histopathological analysis, was completely prevented by zebra
blenny proteins in rats (Ktari etal. 2015). Although saithe
hydrolysate significantly increased the plasma BA (not the
hepatic levels of BA), fecal BA excretion was greatest in the
soy protein fed mice (Liaset et al. 2009). Soy in another
study presented better hypolipidemic effects than the fish
biopeptides. Soy (M/G: 0.24, L/A: 0.79) and salmon frame
proteins (M/G: 0.18, L/A: 1.05) had relatively comparable
effects, with soy diet being more potent over the other, on
hepatic lipid metabolism, fatty acid composition and desat-
urases; reduced 18:1(n-9) to 18:0 ratio (increased hepatic
18:0 (decreased 18:1(n-9)), unchanged Δ9 desaturase gene
expression), reduced 18:3(n-6):18:2(n-6) and
20:4(n-6):20:3(n-6) ratios (reduced mRNA levels of Δ5 and
Δ6 desaturase (reduced activities of Δ5 and Δ6 desaturases)),
increased 20:4(n-6):18:2(n-6) ratio (increased Δ5 desaturase
activity), increased hepatic triacylglycerol-rich lipoprotein
secretion (increased hepatic lipid content), increased hepatic
22:6(n-3) (upregulated peroxisomal acyl-coenzyme A oxidase
with no effect on CPT-1 enzyme activity), and increased
cytosolic isoform of acetyl-CoA carboxylase (increased activ-
ity of lipogenic enzyme fatty acid synthase) compared to
casein diet (M/G: 1.17, L/A: 1.85, negligible health benefiting
effects) in genetically obese Zucker fa/fa rats (Wergedahl
et al. 2004).
Mixed fish diet also could beneficially affect the hyper-
lipidemia in animal models. Concentrations of plasma apo-
lipoproteins were altered by commercial fish protein
preparation compared to casein diet in spontaneously hyper-
tensive rats (SHR); HDL3 apo A-I (-38%), A-II (-55%) and
A-IV (-41%), HDL2 apo A-II (+71%) and A-IV (+41%),
VLDL apo B-48 ( −40%) and apo C ( +40%) (D Ait
Yahia et al. 2005). Total lipid, tissue lipoprotein lipase (LPL)
and hepatic triglyceride lipase (HTGL) activities were sim-
ilarly modulated in fish protein fed SHR compared to casein
diet (liver TL (-24%), and HTGL (-10%), heart TL (+14%)
and LPL (+5%), kidney TL (-6%) and LPL (+24%), gastroc-
nemius TL (-29%) and LPL (+7%), and adipose tissue TL
(-26%) and LPL (-91%)) (D Ait Yahia et al. 2005). The same
commercial fish protein preparation by Seah International
beneficially modulated fatty acid composition of microsomal
lipids, but not casein diet, in SHR animal models; 16:0
(+26%), 18:0 (+21%), 18:1(ω-9) (+20%), 20:4(ω-6) (-50%),
Saturated Fatty Acid (SFA) (+23%), total ω-6 (-29%), and
20:4(ω-6)/18:2(ω-6) (-50%) (Ait-Yahia et al. 2003; D Ait
Yahia et al. 2005).
Clinical studies
Among all fish types, cod was the type mostly tested in
human intervention studies for its anti-hyperlipidemic effect
(Table 5). Different meal sizes and or treatment dosages of
cod protein, compared to various control diets and or pla-
cebo, beneficially affected the lipid peroxidation, oxidative
stress expressed as malondialdehyde/plasma antioxidant
potential (MDA/AOP) ratio, plasma lipid parameters and
antioxidant capacity in overweight, insulin-resistant, diabetic
subjects and or healthy individuals (Gunnarsdottir et al.
2008; Hovland et al. 2019; Mortensen et al. 2009; Ouellet
etal. 2007; Ouellet et al. 2008; Parra et al. 2007; Thorsdottir
et al. 2007; Vikøren et al. 2013). Positive health effects of
control diet at times surpassed the experimental fish diets.
Accordingly, cod test meal, compared to casein, may have
positively influenced fat-induced postprandial lipemia but
its effect was not as significant as whey diet in T2D human
subjects (Mortensen etal. 2009). Mixed fish meal (lean and
or fatty fish) in controlled human interventions modulated
plasma phospholipids (PL), TL, TG, cholesterol, total SFA,
FFA and prostaglandin I2 (PGI2) levels in different types
of population (e.g., coronary heart patients, diabetics with
and or without hypertension as well as healthy subjects)
(Eli K Aadland etal. 2016; Eli Kristin Aadland et al. 2015;
Cui-Feng et al. 2010; de Mello et al. 2009; Erkkilä et al.
2014; Grieger, Miller, and Cobiac 2014; Zaribaf etal. 2014;
C.-F. Zhu, Li, Peng, Li, etal. 2010). Collagenous hydrolysate
preparation, dosage of 13 g daily, could decrease TG, TC,
LDL-c and FFA while increasing PGI2 and HDL-c in T2D
patients (C.-F. Zhu, Li, Peng, Li, et al. 2010). In a more
sophisticated study, fish gelatinous diet supplemented with
or without n-3 PUFA showed hypolipidemic yet rather
sex-dependent activity in free-living insulin-resistant men
and women (Picard-Deland etal. 2012).
Anti-obesity peptides
In the following two sub-sections, effects of fish and
fish-derived products on appetite suppression and regulation
of CCK release, total and organ weight gain/loss including
lipid droplet formation and accumulation as well as food
intake will be discussed in detail.
In-vitro and in-vivo studies
Not only that fish biopeptides have shown to therapeutically
affect the obesity associated comorbidities but also could
directly modulate weight gain and loss, food intake and
energy expenditure. Of all the studies reviewed herein, inves-
tigating anti-weight gain and anti-obesity attributes of
32 S. ABACHI ETAL.
biopeptides, about 44% utilized fish byproducts (about 19
out of 43) (Tables 3–5). Among so many conventional bio-
activity screening studies, Liu et al. attempted a different
approach using response surface methodology for optimizing
the reaction parameter of DH and predicting the anti-obesity
activity of Crucian carp (Carassius carassius) muscle hydro-
lysates in terms of porcine pancreas lipase (PPL) and
α-amylase inhibition effects (L. Liu etal. 2013). Many types
of fish such as Alaska pollock, tuna, blue whiting, smooth
hound, saithe, and others demonstrated anti-obesity activity,
yet cod, salmon and sardine were the most studied ones.
Cod fillet protein, in-house and or commercial preparation,
modulated CCK release, body weight gain (lean and fat
mass), liver weight and fat mass, final weight, food intake,
feed efficiency and growth in multiple in-vitro and in-vivo
experimental setting (Dort et al. 2012; Drotningsvik et al.
2015; Geraedts et al. 2011; Hosomi et al. 2017; Myrmel
et al. 2019). CCK is a hormone that among its so many
major roles, such as stimulation of pancreatic enzyme secre-
tion and growth, could also suppress appetite inducing sati-
ety via CCK1 receptors thus being significant to weight
control (Rehfeld 2017). Outcome of the studies greatly
depended on the experimental setting and the animal model.
Following a monthly 5% cod diet body weight, growth per-
centage and liver weight increased in obese Zucker fa/fa
rats (Drotningsvik et al. 2015). Accordingly, a diet of 23%
cod protein over a month, compared to tuna, increased the
weight gain, final weight, food intake in hypercholesterol-
emic diet fed rats (Hosomi etal. 2017). However, a 20 40%
cod protein intake decreased further weight and fat mass
gain as well as feed efficiency while increasing the lean
mass over 6 12 weeks in high-fat/protein fed obese mice
and low-fat fed lean C57BL/6J mice (Myrmel et al. 2019).
Furthermore, salmon with its anti-obesity effects changed
the mean growth, weight gain, feed intake as well as adi-
ponectin, leptin and resistin levels in rats and mice
(Drotningsvik etal. 2016; Pilon etal. 2011; Vik etal. 2015;
C. Zhu, Zhang, Mu, et al. 2017). Compared to 20% (w/w)
bonito, herring and mackerel protein diets only 20% (w/w)
salmon could over the period of 28-days reduce body weight
gain and visceral adiposity while increasing energy expen-
diture in high fat high sucrose (HFHS) diet fed rats (Pilon
et al. 2011). In the study of Vik et al., while 5% salmon
protein diet increased the feed intake but also prevented
body and total weight gain compared to casein in high fat
fed rats (Vik etal. 2015). Sardine more or less had similar
effects as salmon and cod protein diets. Sardine protein
averted high fructose diet induced weight gain, considerably
in liver and slightly in muscle tissues compared with casein
diet (Madani etal. 2015). Even though sardine protein diet
(with or without high fructose) effects were non-significant
on the weights of heart, kidney and skeletal muscle tissues
but liver weight was much smaller compared to high fruc-
tose diet only (Madani etal. 2015). High fructose fed rats,
supplemented with sardine muscle lysate with L/A ratio of
1.71 (16% higher L/A ratio than cod protein), concomitant
with lower food and energy intakes, gained less weight
which was more prominent in the liver tissue (Madani etal.
2015; Vikøren et al. 2013). Sardine byproduct hydrolysate
was by more than 50% more anti-obesogenic than its muscle
proteins on food intake, body weight gain, food efficiency
rate and fecal lipid excretion (Affane etal. 2018). Interestingly,
fish like saithe had tissue-specific anti-obesogenic effects.
Saithe fish fed rats (taurine and glycine rich diet, M/G:
0.20) gained less weight, pertinent in perirenal and retro-
peritoneal visceral adipose tissues but not in skeletal muscle
(gastrocnemius and plantaris) and hepatic tissues compared
to casein (taurine deficient, low glycine, M/G: 1.08) and soy
(taurine deficient, medium glycine, M/G: 0.24) diets, how-
ever, effects did not root from lower energy absorption
(Liaset etal. 2009). Diet of 20% mixed fish protein (a com-
mercial preparation) reduced absolute liver weight and body
weight gain as well as final body weight, compared with
casein, over 2-months in SHR rats (Ait-Yahia et al. 2003;
Dalila Ait Yahia et al. 2003; D Ait Yahia etal. 2005).
Final body weight of the rats fed fish collagen supple-
mented high caloric diet was +2.2-fold while hypercaloric
and normocaloric fed ones encountered 2.7- and 1.9-folds
weight increase (Raksha etal. 2018). Collagen rich in-house
prepared tuna skin product, at concentration as low as 1 mg
mL−1, prevented the accumulation of lipids and formation
of lipid droplets delaying adipogenic differentiation of pread-
ipocytes while downregulating the expression of adipogenic
(CCAAT/enhancer binding protein alpha (C/EBP) and
PPAR-γ) and target genes (adipocyte protein 2 (aP2)) in
3T3-L1 preadipocytes (E. J. Lee et al. 2017). Same tuna
collagenous preparation substantially reduced the
palmitate-induced lipogenesis in HepG2 cells (E. J. Lee etal.
2017). These health effects were further confirmed in obese
high fat diet fed rats where body weight gain, epididymal
adipocyte size and the expression of adipogenic genes and
its transcription factors were significantly reduced and
downregulated (E. J. Lee et al. 2017). Another commercial
collagenous preparation of skate origin exerted comparable
effects to tuna collagens in obese animal models reducing
liver, visceral and subcutaneous adipose tissue weight (but
not epididymis adipose tissue), lipid droplet size as well as
liver lipid accumulation increasing the adiponectin levels
(Woo etal. 2018). Level of adiponectin with insulin-sensitizing,
anti-inflammatory, and antiapoptotic effects are low in obese,
T2D and at CVD risk patients thus its stimulation may
reduce the prevalence of these conditions by enhancing
metabolism and uptake of glucose (Combs et al. 2001;
Guenther et al. 2014; X. Wu et al. 2003). Anti-obesogenic
activity of glycine rich marine fish collagenic preparation,
concomitant with hypoglycemic effects, may be due to CCK
modulation thus stimulating satiation (Raksha et al. 2018).
Amino acid treatment of 3T3-L1 adipocytes have previously
shown to affect the adiponectin secretion probably by acting
as substrates for the synthesis of fat cell-derived hormone
(Blümer et al. 2008).
It is of note that fish diets, whether lean- and or fatty-fish,
have not always been successful in weight gain prevention.
Like sardine, salmon diet did not change the weight of all
the tested tissues (e.g., epididymal, retroperitoneal, inguinal,
or BAT) and its beneficial effects were only significant in
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 33
the liver weight gain of obese mice (Chevrier et al. 2015;
Madani etal. 2015). Weight gain and fat tissue accumulation
of obese rats were not affected by cod diet and its
anti-inflammatory effects on TNF-α expression in muscle
and adipose tissues were insignificant (Lavigne etal. 2001).
In agreement, Alaska pollock only exerted hypolipidemic
effect with null activity on the weight gain of KK-Ay rats
(Hosomi etal. 2013; Hosomi etal. 2009; Maeda et al. 2015).
Clinical studies
Various fish diets, blue whiting, cod, salmon, and tuna as
well as mixed fish, were intervened in human studies to
further test their clinical anti-obesogenic effects (Table 5).
Fish collagenous preparation of mixed fish and cod, in addi-
tion to anti-obesity effects in rats, exerted stimulatory effects
on the adiponectin levels in T2D (with/without hyperten-
sion) or overweight subjects (Cui-Feng etal. 2010; Hovland
et al. 2019; C.-F. Zhu, Li, Peng, Li, et al. 2010). Cod pre-
sented significant anti-obesity effects in different studies by
means of reducing body weight and waist circumference,
obesity, hunger, prospective foodstuff consumption, energy
intake at the evening meal while increasing satiety and
fasting adiponectin level in healthy overweight/obese subjects
(Borzoei et al. 2006; Gunnarsdottir et al. 2008; Hovland
etal. 2019; Ramel, Jonsdottir, etal. 2009; Thorsdottir et al.
2007). While during the first 4-weeks consumption of cod
protein (with L/A ratio of 1.47) weight and muscle gain
were boosted, fat gain was significantly eased in overweight
human subjects (Vikøren et al. 2013). Blue whiting muscle
hydrolysate in tablet dosage form, at concentrations as low
as 1.4 2.0 g daily, significantly reduced the desire to sweet
consumption before and after experimental diet, weight and
fat mass, body mass index (BMI), extracellular water as well
as waist, hip and thigh circumference while increasing CCK
level in overweight individuals (Nobile et al. 2016; Zaïr
et al. 2014).
Multifaceted character of sh biopeptides toward
prevention and or treatment of MetS
Fish anti-diabetic, anti-obesogenic and anti-hyperlipidemic
peptides exert their beneficial effect on metabolic disorders
non-cytotoxically. Effects of fish anti-MetS peptides are
insignificant on hematological parameters (e.g., white blood
cell (WBC), red blood cell (RBC), total hemoglobin (Hb),
hematocrit (Htc), mean corpuscular volume (MCV), mean
corpuscular hemoglobin (MCH), and mean corpuscular
hemoglobin concentration (MCHC), platelet count (PLT),
and lymph) making them appropriate enough as therapeutic
options for the treatment of MetS (Cudennec et al. 2008;
Y. M. Kim et al. 2015; E. J. Lee et al. 2017; Nasri et al.
2015). Among the studies screening fish peptides for their
anti-diabetic, anti-obesogenic and anti-hyperlipidemic effects,
many have shown to be multi-functional of which few
demonstrated anti-CVD activities. Bogue protein while being
hypolipidemic could also reduce atherosclerotic plaques pre-
venting the formation of foam cells in hypercholesterolemic
diet fed rats (Lassoued et al. 2014). Sardine peptides had
rather similar effects in hypercaloric diet fed rats reducing
plasma AI, CRI, hypertrophy of cells in infarction of heart
tissue, lipid deposition within intima of aorta and lesion
development in aortic wall (Jemil, Abdelhedi, et al. 2017).
Salmon extracted collagen exerted anti-CVD activity and
prevented endothelial thinning and exudative inflammation
in carotid-artery vascular endothelial cells of dietary-induced
diabetic rats (C. Zhu, Zhang, Mu, et al. 2017). Lean fish
meal in 8-weeks intervention study significantly reduced
soluble ICAM-1 (sICAM-1), a biomarker of CVD, in CHD
patients (de Mello et al. 2009). Of the total peptides from
Atlantic herring (Clupea harengus L.) skin and residual
material byproducts proteinic extracts, identified by a com-
bination of MS (Orbitrap), bioinformatics and database
(BioPepDB and Teleost) search, about half were related to
motifs with possible effects on cardiovascular system (EL,
AFL, AIYK+, ALEP, ALPM, APL, AVF, FAL, FY, GPL, HHL,
HLP, IAE, IAP, IAPG, IHPF, IKP, IIAEK+, IKW, IPP, IPY,
IVVE+, IW, KFYG, LGP, LKP, LQGMP, LRP, NIPP, PLPLL,
PPK, PSYVAF, VIKP, VIY, VK, VPP, YN, YNKL, YP, YPK,
YQEP+, YQY) (Pampanin et al. 2012). Furthermore, histo-
pathological analysis has shown that fish bioactive peptides
could preserve kidneys and liver from MetS-associated organ
damages. Hypoglycemic, hypolipidemic, antioxidant and
anti-obesity goby fish hydrolysate prevented renal and
hepatic damages by improving kidney and liver architecture
in hypercaloric fed mice (Nasri et al. 2018; Nasri et al.
2015). Sardine anti-diabetic and antioxidant peptides likewise
decreased renal glomerular atrophy and prevented hepatic
tissue damage in diabetic and oxidative stressed animals
(Jemil, Nasri, etal. 2017). Hypoglycemic and hypolipidemic
zebra blenny hydrolysates exerted comparable effects as sar-
dine and goby fish in diabetic and hypercaloric diet fed rats
(Ktari etal. 2015; Ktari etal. 2013). Another example could
be mackerel hydrolysate peptides demonstrating significant
in-vitro hypotensive and immunoregulatory effects with neg-
ligible in-vivo anti-diabetes and anti-obesity attributes at the
tested concentrations (Soheila Abachi et al. 2022;
Hokmabadinazhad etal. 2022). Many fish-extracted peptides
with dual- and multi-functional activities in animal and
human studies have been reviewed in this work (Tables
4 and 5).
Nonetheless, MetS benefiting health effects (glucose and
lipid metabolism) of the fish diets are rather divergent
depending on the characteristics of the under study popu-
lation and study design (Dale et al. 2018; Hovland et al.
2019). Various marine fish collagenous preparations had
affected T2D patients, with and or without high blood pres-
sure, in different human intervention study settings
(Cui-Feng etal. 2010; C.-F. Zhu, Li, Peng, Li, etal. 2010).
Serum levels of hsCRP, a metabolic regulator, and adipokine
(leptin and resistin) were beneficially modulated in the study
of Cui-Feng etal. by a marine collagen product yet some-
what similar preparation exerted null effect on the same
parameters in a similar randomized double-blind study of
Zhu et al. (Cui-Feng et al. 2010; C.-F. Zhu, Li, Peng, Li,
et al. 2010). Various fish diets (e.g., tuna and cod) reduced
34 S. ABACHI ETAL.
MetS (same criteria defined by health organizations) in over-
weight and obese subjects (supplementary Table 1) (Ramel,
Jonsdottir, et al. 2009; Zaribaf et al. 2014). Logically, meal
and or dosage size were important in the health effects
hence fish ought to be ingested in adequate amounts to
exert significant beneficial therapeutical activity. Accordingly,
in the study of Grieger etal., a 2-months low fish diet could
not significantly modulate the cardiovascular biomarkers in
a healthy elderly population (Grieger, Miller, and
Cobiac 2014).
Structure activity relationship and stability
Scientific community agrees on the fact that certain features
of the bioactive material play vital in their positive biological
activity. For instance, fish hypotensive and immunomodu-
latory peptides commonly possessed smaller molecular
weight, short chain, and certain compositional factors yet
presence or absence of a feature did not necessitate its effec-
tiveness and or ineffectiveness. The topics have been thor-
oughly reviewed by the authors. More or less same applies
to anti-diabetes, anti-hyperlipidemia, and anti-obesity fish
biopeptides as discussed in the following paragraphs. For
example, food-derived oligopeptides of small molecular
weight and shorter amino acid sequences have repetitively
been reported as effective DPP-IV inhibitors (R. Liu, Cheng,
and Wu 2019). Among numerous reported food-derived
DPP-IV inhibitors, up to 2019, more than 88.4% have MW
smaller than 1,000 Da, and around 47% are hydrophobic
indicating the importance of physicochemical properties of
the biopeptides in their bioavailability and bioefficacy (R.
Liu, Cheng, and Wu 2019). Both hydrophobic and hydro-
philic peptides could inhibit the activity of DPP-IV although
chance of a hydrophobic peptide (hydrophobicity index of
larger than 0) being an anti-DPP-IV is higher (Nongonierma
and FitzGerald 2013). Presence of N-terminus HAA and or
aromatic AA (AAA) has been a good indication of a food
derived anti-DPP-IV peptide yet other features also come
into play for successful inhibition of the enzymes activity
(Nongonierma and FitzGerald 2014). In addition to molec-
ular mass and hydrophobicity, presence and or absence of
particular amino acids in certain positions of the sequences
seem to fully and or partially contribute to the anti-MetS
activity of fish biopeptides. Anti-MetS affecting features of
peptides have been evidently documented in many studies
by comparing the experimental peptide and or diet to ref-
erence proteins such as casein (supplementary Table 4).
Occurrence of proline as N-terminus penultimate residue
along with HAA as N-terminus ultimate residue seems to
be rather important characteristics of a food-originated
DPP-IV inhibitor (R. Liu, Cheng, and Wu 2019). In an
in-silico study aiming to predict food derived DPP-IV inhib-
itors, potential candidate peptides had N-terminal trypto-
phan with proline at penultimate position (Nongonierma
and FitzGerald 2014). Therefore, in addition to the occur-
rence of proline at the penultimate point of N-terminal,
presence of HAA at the C-terminal and N-terminal ultimate
position appears to facilitate the glucoregulatory effect of a
biopeptide (R. Liu, Cheng, and Wu 2019; Matsui, Oki, and
Osajima 1999). N-terminal tryptophan of barbel-extracted
tri-peptide, WSG, also played significant in its anti-DPP-IV
activity (Assaad Sila et al. 2016). While whey isolated LLF
and LV were impotent anti-DPP-IV peptides, LL and LA
potently inhibited the activity of the same enzyme noting
the significance of N-terminus position 3 residue along with
ultimate and penultimate motifs (Tulipano et al. 2011). In
the study of Huang etal., N-terminal proline was as import-
ant as N-terminal penultimate proline for the DPP-IV inhi-
bition (S.-L. Huang et al. 2012). Interestingly, replacement
of leucine with tyrosine in the same YPL sequence, tyrosine
at both N- and C-terminals, decreased the potency by about
7-fold, from IC50 of 3.9 mM to 25.8 mM (S.-L. Huang etal.
2012). In-vitro DPP-IV inhibition of 3 mg mL−1 tilapia and
milkfish skin gelatin hydrolysates, with a higher amount of
imino acids, were by about 8% greater than halibut and
hake skin gelatin hydrolysates (T.-Y. Wang et al. 2015). In
agreement, proline or hydroxyproline (a metabolite of pro-
line) supplementation has shown to enhance the perfor-
mance and weight gain of vertebrates and invertebrates (G.
Wu etal. 2011). Accordingly, proline and HAA are present
at the same positions as of salmon, halibut and tilapia oli-
gopeptides (Li-Chan et al. 2012; T.-Y. Wang et al. 2015).
Glycine in free form, as a supplement, has demonstrated
many health effects (e.g., anti-inflammation, immunomod-
ulation, cytoprotection, etc.) through glycine-gated chloride
channel activation in various cell types in the in-vivo exper-
iments. Glycine exerts various cardioprotective and vascular
health effects such as GLP-1 and glucagon secretion stim-
ulation, downregulation of Kupffer cell activation controlling
fructose-induced steatosis, and sucrose-induced MetS inhi-
bition (Table 1). This may be the reason for the glucoreg-
ulatory, anti-hyperlipidemia and anti-obesogenic activities
of glycine rich tilapia, salmon, flathead gray mullet and
marine fish collagenic preparations and hydrolysates (H.-S.
Kim, Nian, et al. 2009; Raksha et al. 2018; J. Wang et al.
2010; R. Zhang, Chen, Chen, et al. 2016; C.-F. Zhu, Zhu,
and Zhou 2013; C. Zhu, Zhang, Mu, et al. 2017). Glycine
and sulfur-containing amino acids (methionine, cysteine,
homocysteine, and taurine) have long been known as poten-
tial lipid metabolism modulators, increasing HDL-c and
decreasing VLDL-c, at appropriate concentrations conceiv-
ably via [1] altering the enzymatic and transcriptional activ-
ity by reduced form of glutathione and stimulating cholesterol
degradation and its clearance from blood circulation, [2]
changing the posttranslational modification pattern of ligand
binding to nuclear receptors and phosphorylation/dephos-
phorylation regulatory proteins, and [3] inducing CYP7A1
and Apo A-I gene expressions (Oda 2006). However, diets
of varying richness and deficiency of glycine and or M/G
as well as taurine behave differently on weight gain, specif-
ically adipose tissue, and not skeletal muscle, of rats.
Correspondingly, taurine and glycine rich saithe fish (rather
balanced M/G) was more anti-obesogenic than casein and
soy (Liaset et al. 2009). Higher amounts of these amino
acids in saithe hydrolysate also enhanced the hepatic secre-
tion of BA and its consequent excretion into plasma which
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 35
seemed to be more affected by glycine than taurine since
organic anion transporting polypeptide (OATP) 1, bile acid
coenzyme A: amino acid N-acyltransferase (BAAT), and
ABC subfamily C member 3 (ABCC3) hepatic expressions
were not modulated by taurine-rich diet (downregulating
hepatic ABCC4 protein levels, hepatic expression of, OATP1
and OATP4 significant in cholestasis) (Liaset et al. 2009).
Expression of BA transporters and BA fecal secretion have
been shown to be sensitive to taurine and glycine of the
treatment diet (Liaset etal. 2009). While higher fecal excre-
tion of BA in soy diet has been theorized to be due to
irreversible binding of its indigestible fractions to BA, higher
plasma BA in saithe diet fed rats may be because of taurine
and glycine making the conjugation of BA possible hence
improving their solubility and liver secretion to the blood
(Liaset et al. 2009). According to Iwami et al.,
anti-hypercholesteremic peptides in part owe their effectivity
to the enhanced binding of hydrophobic plant biopeptides
to BA in the digestive tract (Iwami, Sakakibara, and Ibuki
1986). In the study of Liaset et al., hepatic expression of
OATP1, BAAT and ABCC3 weren’t as much affected by
taurine, however, overall those may have been sensitive to
glycine (Liaset et al. 2009). Interestingly, taurine deficient
(casein with low glycine) and high taurine (saithe with high
glycine) diets downregulated the hepatic expression of
OATP1 (Liaset et al. 2009). In contrary, hepatic expression
of BAAT and ABCC3 were upregulated by soy (taurine
deficient, medium glycine) and saithe (taurine and glycine
rich) diets (Liaset etal. 2009). Beneficial effects of fish diet
could as well possibly be explained by its satiety attributes
over chicken and beef intake which have been related to
the presence and absence of certain amino acids such as
taurine (Uhe, Collier, and O’Dea 1992). Amino acid com-
position of a satiating fish diet was relatively different from
beef and chicken (fish: CAA 27 > HAA 23 > BCAA 11 > Tau
1.5 g 50 g−1 protein, L/A 1.69 and M/G 0.53, chicken: CAA
20, HAA 20 > BCAA 10 > Tau 0.2 g 50 g−1 protein, L/A 1.74
and M/G 0.60, beef: CAA 24 > HAA 22 > BCAA 11 > Tau
0.2 g 50 g−1 protein, L/A 1.59 and M/G 0.25) with relatively
high CAA, high taurine, balanced L/A and M/G ratios (Uhe,
Collier, and O’Dea 1992). Beef, pork, veal, eggs, milk, and
milk products (BPVEM) diet was to a small fraction richer
in BCAA, HAA, and EAA, up to 11%, with higher L/A
ratio (7%) compared to cod diet with stimulatory effect on
insulin sensitivity in human subjects (Ouellet et al. 2007).
The importance of L/A ratio peptides was pointed out some
decades ago by Kritchevsky et al. demonstrating the ath-
erogenicity power of different diets with parallel lysine but
variable arginine levels. Diet with highest arginine thus low-
est L/A presented the least atherogenicity in rabbits
(Kritchevsky etal. 1982). Typically, low M/G and L/A ratio
diets have repeatedly demonstrated anti-atherogenic,
anti-hyperlipidemic and anti-obesogenic effects in many
studies and various experimental settings (Gudbrandsen
etal. 2005; Kern etal. 2002; Morita etal. 1997; Rajamohan
and Kurup 1990; Vega-López et al. 2010). In a study, bal-
anced L/A (1.1) diet fed cobia performed better than high
(1.8) and low (0.8) L/A diet fed fish (Van Nguyen et al.
2014). Overall, weight gain of cobia (lipid and protein gain)
were lower in imbalanced L/A diet fed fish (Van Nguyen
etal. 2014). Similar finding was observed in a human inter-
vention study where L/A and M/G ratios of casein/whey
diet (EAA 1255 > HAA 1201 > CAA 1183 > BCAA 587 > imino
acids 262 mg d−1) were considerably higher (2.41, 1.53) than
anti-diabetic fish protein diets (Hovland et al. 2019). Yet,
zebra blenny may among other assumptions, owe its hypo-
glycemic, antihyperlipidemic and other possible health effects
to its high BCAA and CAA mainly leucine (15%), glutamic
acid and arginine (20%), approximately accounting for 35%
of the total (Ktari etal. 2013). It is of note that oral sup-
plementation of arginine in free form, a CAA, to acute
myocardial infarction patients with 3 g of the amino acid
daily improved their dyslipidemia conditions, decreasing
serum TC and TG while increasing HDL-c, during the
2-weeks period of study (Tripathi, Misra, and Pandey 2012).
It should be noted that along with beneficial effects there
are various examples of proline, charged and aromatic amino
acids having negative effect on the glucoregulatory attributes
of fish biopeptides and same have been valid for
anti-hyperlipidemic activities of fish proteins.
Leucine, comparable to glycine with glucoregulatory
effects, has been known for its stimulatory effect on pan-
creatic insulin secretion via activation of mammalian target
of rapamycin (mTOR) pathway successively downregulating
the surface expression of adrenergic α2A receptor and
enhancing cAMP production (Table 1) (Yang et al. 2012).
In addition, leucine, in free form, like several other amino
acids, tended to modulate MetS-induced insulin resistance,
hepatic steatosis reversing the inflammation of adipose tissue
over 2-months consumption of dietary supplements in obese
rats (Macotela et al. 2011). Presence of leucine at the
C-terminal and tyrosine at the N-terminal, contrary to
DPP-IV inhibitors, have been reported vital for the inter-
action of a biopeptide with active binding sites of
α-glucosidase attenuating its activity (Yu Zhang, Chen, Chen,
et al. 2016). Collectively, molecular weight, chain length,
compositional and structural features of a fish biopeptide
are important for anti-MetS effects. Additionally, amino acid
composition and their respective bioactivities may vary
depending on the starting material and type of fish. For
instance, white muscle of cod tended to be a relatively more
potent hypocholesterolemic protein than tuna red muscle
protein which may be due to compositional differences of
diets (Hosomi et al. 2017).
Structural dierences between dierent sh and
control diets aecting their bioactivities
In the following sub-sections, authors have attempted to
focus on the compositional differences of biopeptides based
on most investigated fish types, where possible, for better
understanding of the structure-activity relationship.
Alaska pollock
Importance of either extremities of a biopeptide in gluco-
regulatory activity of fish hydrolysate was examined by frag-
menting an Alaska pollack-originated (myosin(548–556))
36 S. ABACHI ETAL.
oligopeptide, ANGEVAQWR, to tri-peptides; ANG, EVA,
and QWR (Ayabe et al. 2015). Interestingly, at 1 mg kg−1
C-terminal tri-peptide QWR had glucose lowering effect in
diabetic rats while its di-peptide fragments (QW, and WR)
were ineffective (Ayabe et al. 2015). Another C-terminus
Alaska pollack-originated pentapeptide, YNELR, and
tri-peptide, ELR, tended to lower the blood glucose levels
at the dosage of 1 mg kg−1 in animal models, while the main
identified actin(87–97) associated oligopeptide
(IWHHTFYNELR) had negligible effects (Ayabe etal. 2015).
Glucose uptake rates in mouse skeletal muscle C2C12 cells
were tested to further elucidate the structure activity rela-
tionship of fish glucoregulatory peptides (Ayabe etal. 2015).
Tripeptides, QWR, ELR and pentapeptide, YNELR, enhanced
the uptake of glucose in cells (Ayabe et al. 2015). At con-
centrations up to 500 μM QWR dose-dependently activated
the insulin-independent glucose uptake in skeletal muscle
cells (Ayabe etal. 2015). Potency of Alaska pollock hydro-
lysate may be related to glycine content of the diet as it
composed of 2-fold glycine compared to casein yet suppos-
edly both diets were taurine deficient (not listed in AA
composition of diets) (Hosomi et al. 2012). Casein with
ineffective attributes on hepatic TG and FA desaturase indi-
ces of obese T2D KK-Ay mice model compared to Alaska
pollock muscle proteins has higher proline (+3.2-times), yet
lower glycine (-0.47-times) (Maeda etal. 2015). Briefly, pres-
ence of arginine at C-terminus and polar amino acids at
N-terminus position as well as high glycine or less proline
content may play significant in bioactivity of Alaska pollock
oligopeptides on glucose uptake and level.
Salmon
Amino acid compositions of salmon spine and backbone/
head diets, regardless of molecular weight distribution, were
to some extent comparable, yet their beneficial metabolic
attributes greatly differed from one another (Vik etal. 2015).
Backbone lysate of salmon contained least of BCAA and
CAA, whilst was richest in imino acids with least M/G and
L/A (0.35 and 1.13, respectively) (Vik et al. 2015).
Accordingly, inadequate sub-optimal methionine diet fed
animal model (Atlantic salmon) compared to adequate opti-
mal amount fed ones, led to increased liver weight, FAS
activity, 18:1/18:0 FA ratio, hepatic TG concomitant with
decreased hepatic taurine and total fecal and plasma BA
(descriptive of nonalcoholic fatty liver disease development
in rats) (Espe etal. 2010). For example, umamizyme salmon
spine lysate, with rather high M/G, tended to be hyperlip-
idemic than being hypolipidemic (Vik et al. 2015). Salmon
hydrolysate, compared to herring, in the study of
Drotningsvik etal., presented different patterns of bioactivity
on MetS markers being more hypoglycemic and
anti-obesogenic than hypolipidemic in an obese and diabetic
animal model (Zucker fa/fa rat) (Drotningsvik etal. 2016).
Salmon and herring byproduct hydrolysates contained com-
parably similar amounts of HAA, CAA and BCAA yet their
taurine quantities, L/A and M/G ratios significantly diverged
from one another and from the casein/whey control diet
(taurine deficient) (Drotningsvik et al. 2016). Of the
nineteen tested hypocholesterolemic and/or antidiabetic
sequences (ALPMH, GGV, GLDIQK, GPAE, GPGA, GPL,
HIRL, IAVPGEVA, IIAEK, IPI, LPGP, LPYPR, PGPL,
RPLKPW, VAWWMY, VGVI, VGVL, VPDPR, VVYP, VW,
VYVEELKPTPEGDLEILLQK, YPFVV, and YYPL) only few
were present in the herring and salmon hydrolysates (her-
ring: GPL, IPI, VW, GPAE and LGPG (antidiabetic motifs),
IIAEK (hypocholesterolemic motif), PGPL (hypocholester-
olemic and antidiabetic motif), salmon: GPL, IPI and VW
(antidiabetic motifs), PGPL (hypocholesterolemic and anti-
diabetic motif)) (Drotningsvik et al. 2016). In conclusion,
type of salmon as well as extraction technique and/or type
of enzyme seem to be effectual on its anti-MetS attributes.
Moreover, presumably adequate taurine level as well as M/G
and L/A ratios define the bioactivity of salmon
biopeptides.
Cod
Specific amino acid arrangement of cod may be the source
of its metabolic health effects on insulin secretion or insulin
excretion rates in the liver (von Post-Skagegård, Vessby, and
Karlström 2006). Glucoregulatory effects of cod muscle pro-
teins compared to soy and casein-fed animals in the study
of Lavigne etal., on glucose and insulin dynamics, may be
explained by its balanced L/A and M/G ratios (1.50 and
0.37, respectively) (Lavigne, Marette, and Jacques 2000). The
amino acid composition of all three diets were comparable
except that casein contained significantly higher amounts
of proline (1.9-times) compared to cod and soy (Lavigne,
Marette, and Jacques 2000). In another study, comparable
anti-obesogenic effects were observed by similar diets on
high fat fed rats (relatively parallel amino acid composition
of diets with significantly different L/A, M/G ratios) on
obesity-induced muscle insulin resistance and
insulin-stimulated glucose uptake in skeletal muscle cells
(Lavigne etal. 2001). In both of Madani etal. and Chevrier
et al. studies, with insufficient effects of experimental diets
on weight gain, the L/A ratio of casein hydrolysate compared
to salmon and sardine lysates was higher (Chevrier et al.
2015; Madani et al. 2012; Madani et al. 2015). In another
sturdy, casein diet with negligible antidiabetic and
anti-obesogenic effect on obesity prone rats had almost
+4-times more proline than cod/scallop diet (Tastesen et al.
2014). Similar to the studies of Madani etal. and Tastesen
etal., the ineffective casein diet on weight gain, plasma and
liver lipid and cholesterol levels was rich in CAA, BCAA,
AAA and proline (+100%) with higher L/A ratio, yet
glycine-deficient compared to purified fish protein (Ait-Yahia
et al. 2003; D Ait Yahia et al. 2005). L/A and M/G ratios
of diets varied greatly from one another (in the decreasing
order of casein > chicken and > cod/scallop) (Tastesen et al.
2014). This agrees with the study of Van Nguyen et al. in
which balanced L/A ratio fed cobia performed better than
high and low L/A ratio fed fish (Van Nguyen et al. 2014).
It is noteworthy that plasma TC, HDL-c and LDL-c as well
as liver TG and total neutral lipid concentrations, concom-
itant with increased mRNA expression of lipogenic gene
diacylglycerol acyltransferase 1 (DGAT1), were higher in
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 37
chicken diet (with medium taurine) fed rats (Tastesen
et al. 2014).
In accordance, taurine and glycine concentrations could
have possibly been high enough in cod/scallop diet for
beneficial management of lipid metabolism in various
animal experimental models (I.-J. Jensen et al. 2016;
Tastesen et al. 2014). In the study of Tastesen et al.,
glycine and sulfur containing amino acid concentration
of the cod/scallop regimen with anti-obesogenic and glu-
coregulatory activity was relatively different from other
experimental diets (glycine (mmol per kg of diet); cod/
scallop 192 > chicken 109 > casein 47 and sulfur containing
amino acids (mmol per kg of diet); cod/scallop
137 > chicken 85 > casein 70) (Tastesen et al. 2014). A
similar trend was observed in the study of Jensen et al.
testing the metabolic beneficiary effect, mainly on ath-
erosclerotic lesions, of cod/scallop dietary intake over
3-months on high fat fed ApoE−/− rats (mg of glycine per
g of diet); cod/scallop 10.1 > chicken 5.7, and sulfur con-
taining amino acids (mg per g of diet); cod/scallop
13.7 > chicken 8.3) (I.-J. Jensen et al. 2016). It is worthy
of note that taurine has also been documented as an
amino acid with satiating effects which would decrease
food intake while lowering weight gain yet non-hydrolyzed
and hydrolyzed goby fish proteins, taurine-deficient, had
similar effects (Nasri et al. 2018). Single dose of cod
taurine-rich (6.6 mg per g of protein) enzymatic hydro-
lysate, compared to taurine-deficient casein intake with
high L/A and M/G ratios, however, could only depress
postprandial insulin secretion with null effect on blood
glucose response or GLP-1 levels (Dale et al. 2018).
Contrary to oral administration of single cod dose to
middle-aged healthy individuals with slight glucoregula-
tory effect, an 8-weeks mixed low-fish diet only minimally
affected cardiovascular biomarkers in the healthy elderly
population (Grieger, Miller, and Cobiac 2014). Taurine
concentration was significantly higher in cod/scallop with
beneficial effect on HDL-c, 61 mmol kg−1 of diet, than in
chicken and casein ones (Tastesen et al. 2014). Casein
diet, with glucose tolerance reduction effect, had the high-
est BCAA (14 39%) compared to cod/scallop and chicken
diets (Tastesen et al. 2014). Contrary to casein, chicken
with 290 mmol of BCAA, 630 mmol of EAA and 611 mmol
of CAA per kg of diet, boosted the plasma insulin in the
obesity-prone animals (Tastesen et al. 2014). In compar-
ison to chicken diet, cod/scallop had correspondingly by
about up to −15% less CAA, EAA and BCAA with smaller
L/A and M/G ratios (-18% and −44%, respectively), how-
ever, its taurine content was +53-times higher (I.-J. Jensen
etal. 2016). Taurine rich cod/scallop and taurine deficient
casein diets had higher EAA but lower CAA content than
chicken (Tastesen et al. 2014). Following 7-weeks of dif-
ferent protein diet regimens, compared to chicken fed
group, seafood and casein diet decreased the efficiency
of feed and adiposity in obesity-prone rats (Tastesen et al.
2014). In Brief, adequate amount of proline, glycine, tau-
rine, BCAA, CAA and AAA as well as balanced L/A ratio
are positively correlated with anti-MetS properties of cod
biopeptides.
Sardine
Similar compositional factors were important for sardine’s
hypoglycemic, hypolipemic and anti-obesity effects.
Alpha-glucosidase inhibition effectivity of truncated sardine
muscle extracted peptide (YPL) was insignificantly different
from the original tetra-peptide (YYPL) indicating the impor-
tance of YPL sequence for glucoregulatory activity of the
biopeptide (Matsui, Oki, and Osajima 1999). Even though
shorter in length, synthetic di-peptide analogues of the same
sequence, YP and PL, were negligibly potent against
α-glucosidase (Matsui, Oki, and Osajima 1999). Substitution
of Leucin, a HAA, with another hydrophobic one, glycine,
in the Tyr-Pro-Leu sequence slightly decreased its activity
(Matsui, Oki, and Osajima 1999). Both of sardine diets,
hypolipidemic byproduct and fillet proteins, as well as casein
control diet composed of comparable amounts of CAA,
HAA and BCAA, however, byproduct extracted diet con-
tained the highest amount of taurine and glycine while its
proline was lowest of all (-29% and −78% compared to fillet
and casein diets, respectively) (Affane et al. 2018). Taurine
contents of sardine diets were +2.9-times and +1.7-times
higher (sardine byproduct and or fillet respectively) than
that of casein one (Affane etal. 2018). In addition to tau-
rine, the higher efficacy of sardine byproduct diet on hyper-
lipidemia, obesity, atherogenicity and oxidation, may be due
to its lower M/G rate compared to sardine fillet protein and
casein by about −31% and −64%, correspondingly (Affane
et al. 2018). More potent hypolipidemic bogue hydrolysate,
with −25% lower L/A and −56% less BCAA, particularly
was taurine- (+2.46-times) and glycine-rich (+1.33-times)
and had more sulfur containing amino acid (+1.95-times)
in comparison to sardine hydrolysate with slightly smaller
anti-dyslipidemia activity (Benomar et al. 2015). Clinical
oral dose of l-arginine (3 g daily) has exerted antihyperlip-
idemic effects, modulating the lipid profile, in elderly
patients diagnosed with acute myocardial infarction (Tripathi,
Misra, and Pandey 2012). Therefore, superiority of bogue
in lipid metabolism modulation, compared to sardine, may
be due to its high arginine content (1.5-times) (Benomar
et al. 2015). Both fish diets, sardine and bogue, had com-
parable M/G ratios (0.41 and 0.42, respectively) (Benomar
et al. 2015). Amino acid composition of casein, with null
effect on weight gain of obese rats, and sardine slightly
differ from one another. Though abundance of negatively
CAA in both is relatively comparable, casein has +2.5-fold
more proline than sardine and its L/A was higher than
sardine (Madani et al. 2015). In conclusion, presence of
polar amino acids at N-terminal along with branched chain
and/or hydrophobic amino acids at C-terminal influence
anti-MetS activity of sardine oligopeptides. Furthermore,
balanced L/A and M/G ratios plus adequate taurine and
glycine as well as low proline play rather a vital role in the
bioactivity of sardine similar to other types of fish discussed
earlier.
Tilapia
Metabolite of proline, hydroxyproline, seemed to play a
rather significant role on the GLP-1 secretory and
38 S. ABACHI ETAL.
anti-DPP-IV activity of fish biopeptides. Imino acid rich
tilapia and or halibut induced GLP-1 secretion, unlike
salmon gelatinous hydrolysate with insignificant stimulatory
effect on the same incretin, in diabetic rats (Hsieh et al.
2015; T.-Y. Wang et al. 2015) (Hsieh et al. 2015). Tilapia
hydrolysate inhibited plasma DPP-IV activity by 28% where
sitagliptin hindered this activity by 63% in diabetic rats
(T.-Y. Wang etal. 2015). Furthermore, the same hydrolysate
was more efficient than halibut and sitagliptin in stimulat-
ing the secretion of GLP-1 in comparison to the diabetic
control group where the substrates secretion was signifi-
cantly lower than non-diabetic normal animals (T.-Y. Wang
etal. 2015). Tilapia hydrolysate with higher imino acid was
as potent as sitagliptin in enhancing the secretion of insulin
in experimental rats where halibut hydrolysates effect was
marginal (T.-Y. Wang et al. 2015). Both tilapia and halibut
identified sequences contained the well-known GLP-1 secre-
tion stimulatory motifs (LGG, GL and GP) which exert
their effect via proton-coupled uptake and the
calcium-sensing receptor (Diakogiannaki etal. 2013) (T.-Y.
Wang etal. 2015). Halibut- and salmon-originated peptides
also contain glutamine (Q) which has repeatedly been
reported as a strong GLP-1 secretagogue (Reimann et al.
2004) (Harnedy etal. 2018a). Bester sturgeons, halibut, and
salmon oligopeptides contain a well-known anti-DPP-IV
biopeptide (GPAG) while GPA was also a common motif
in many of antidiabetic fish peptides reviewed herein (Hsu
et al. 2013; Sasaoka et al. 2018; T.-Y. Wang et al. 2015; C.
Zhu, Zhang, Mu, et al. 2017). The same tri-peptide, GPA,
has been reported as a DPP-IV inhibitor and the same
motif has occurred in few of the fish anti-DPP-IV biopep-
tides (Bauvois 1988; Hatanaka, Kawakami, and Uraji 2014;
Li-Chan et al. 2012; Neves et al. 2017; Ying Zhang, Chen,
Chen, et al. 2016). In closing, tilapia biopeptides may owe
their anti-diabetic properties to the presence of hydrophobic
amino acids at both extremities.
Herring
In the study of Durand etal., while herring milt hydrolysate
had insignificant stimulatory effect on the glucose uptake,
its Electrodialysis with Ultrafiltration membrane (EDUF)
fraction (non-charged peptides) tended to significantly
increase the in-vitro glucose uptake of skeletal muscle cells
(Durand etal. 2019). In contrary, positively charged peptides
of herring milt hydrolysate decreased the glucose uptake
rate (≤ −9.9%) in the same cells and similar analytical con-
ditions (Durand et al. 2019). Examining the amino acid
composition of the EDUF herring milt fractions, obviously
the most potent one has the least CAA (32 g 100 g−1 of
peptides versus 45 and 49 g 100 g−1 of peptide for cationic
and anionic fractions respectively) while having highest tau-
rine content (1.3 g 100 g−1 of peptide versus 0.58 and 1.13 g
100 g−1 of peptide for cationic and anionic fractions respec-
tively) with rather balanced L/A ratio (0.13 versus 0.10 and
0.29 for cationic and anionic fractions respectively) (Durand
et al. 2019). Not only that taurine harbors hypolipidemic
and satiating properties but also has repeatedly demonstrated
hypoglycemic effects in different experimental platforms and
the topic has been reviewed by Ito etal. (Ito, Schaffer, and
Azuma 2012; Kulakowski and Maturo 1984; Tenner, Zhang,
and Lombardini 2003). Hence, the potent effect of herring
EDUF non-charged fraction may be due to its relatively
higher taurine content rather than the charge of peptides
(Durand et al. 2019). It is noteworthy that the effective
fraction of herring milt hydrolysate and the non-dialyzed
hydrolysate had similar amino acid composition properties
except for its lower taurine (-5%) and higher L/A ratio
(+15%) (Durand et al. 2019). Atlantic herring is rich in
anti-CVD peptide motifs in which alanine at N- and or
penultimate position of N-terminal, proline at C- and or
penultimate point of C-terminal and leucine rarely in
C-terminal and generally at N- or penultimate position of
N-terminal are situated (Pampanin etal. 2012). These obser-
vations demonstrate the fact that certain conformational
structure, chemical, physical, and biochemical properties
affect the bioactivity of fish biopeptides. It could be con-
cluded that adequate amount of charged amino acids and
presence of hydrophobic amino acids at N- or C-terminals
as well as their penultimate positions could explain anti-MetS
effects of tilapia biopeptides. Furthermore, taurine content
of the fish may also be imperative for its biological activities
against MetS.
Stability of peptides to gastrointestinal digestion
Almost all of anti-hyperlipidemic and anti-obesity along
with the majority of anti-diabetic fish biopeptides were
tested in animal studies presenting significant hypoglyce-
mic, hypolipidemic and anti-obesogenic potencies meaning
they could resist the harsh environment of the digestive
tract and be highly bioavailable within the tract of exper-
imental models such as fish, pigs, sheep, rats, and mice.
Human studies intervening fish diets in many different
healthy and or ill subjects are also confirmatory of those
studies demonstrating successful effects against diabetes,
obesity, CVD and MetS (Table 5). In line, few studies
followed the same approach in cell and or in-vitro settings
testing the stability of anti-MetS fish biopeptides in static
and dynamic stimulated gastrointestinal digestion systems.
To reveal the bioavailability of fish hypocholesterolemic
peptides Hosomi et al. lysed Alaska pollock muscle and
casein proteins to soluble and insoluble fractions by diges-
tive proteases and tested BA binding capacity and micellar
solubility of cholesterol (Hosomi et al. 2011). In a simu-
lated intestinal digestion (pepsin and pancreatin hydrolysis)
model, soluble and insoluble fractions of Alaska pollock
proteins tended to reduce the micellar solubility of cho-
lesterol, compared to casein fractions, which may explain
the higher fecal cholesterol excretion rates in rats (Hosomi
et al. 2011). A shark liver peptide (S‐8300) with great
hepatoprotective effects was susceptible to static GI diges-
tion (susceptible to trypsin and proteinase K, resistant to
DNase, RNase, heat, acid and alkali) (F. Huang and Wu
2005, 2010; F. J. Huang & Wu, 2010). Moreover, the same
peptide showed significant anti-MetS activity in animal
studies which may be due to the fact that GI proteolytic
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 39
activity may have lysed the rather big S‐8300 molecule
into more potent smaller size oligopeptides (F. Huang and
Wu 2005, 2010; F. J. Huang & Wu, 2010). Similar approaches
were taken to assess the stability of anti-diabetes
biopeptides.
In another attempt, glucoregulatory effects of salmon
byproduct biopeptides were put to test in a static GI digestion
(pepsin and corolase PP) model (Harnedy etal. 2018a). Skin
gelatin lysates were relatively susceptible to the digestive
enzymes (insulin secretion from BRIN-BD11 cells +3.2- and
+8.0-times pre- and post-digestion, GLP-1 secretion from
enteroendocrine GLUTag cells +3.2- and +0.4-times pre- and
post-digestion, DPP-IV activity IC50 0.90 and 1.19 mg mL−1
pre- and post-digestion) (Harnedy et al. 2018a). However,
trimming hydrolysates rather resisted the GI protease activity
(insulin secretion from BRIN-BD11 cells +3.0- and +4.2-times
pre- and post-digestion, GLP-1 secretion from enteroendo-
crine GLUTag cells +2.4- and +2.3-times pre- and
post-digestion, DPP-IV activity IC50 0.84 and 1.00 mg mL−1
pre- and post-digestion) (Harnedy etal. 2018a). Insulin stim-
ulatory potency of the salmon biopeptides was notably
enhanced by digestion, however, their anti-DPP-IV and
GLP-1 induction activities were reduced or unaltered
(Harnedy et al. 2018a). Hydrolysate of blue whiting muscle
behaved similar in the simulated digestion (pepsin and coro-
lase PP DH: 33%) analysis (insulin secretion from BRIN-BD11
cells +3.8- and +4.2-times pre- and post-digestion, DPP-IV
activity IC50 1.28 and 1.49 mg mL−1 pre- and post-digestion,
membrane depolarization +6.4-times, [Ca2+]i and cAMP con-
centration in BRIN-BD11 cells post-digestion) (Harnedy etal.
2018b). Blue witting hydrolysate was further lysed, to
pre-examine its in-vivo permanence, by the proteolytic activ-
ity of GI enzymes to di- and tri-peptides (<500 Da peptides:
65%) altering the behavior of post-digestion lysate on insulin
secretion, DPP-IV and membrane potential (Harnedy et al.
2018b). In accordance, boarfish GI proteases (pepsin and
corolase PP) subjected sarcoplasmic hydrolysate was fairly
resistant to the digestion (insulin secretion from BRIN-BD11
cells +4.2- and +5.3-times pre- and post-digestion, GLP-1
secretion from enteroendocrine GLUTag cells +1.3-times
post-digestion, DPP-IV activity IC50 1.18 and 1.21 mg mL−1
pre- and post-digestion, glucose uptake in 3T3‐L1 adipocytes
+1.3-times post-digestion) (Parthsarathy etal. 2019). Digestive
enzyme lysed boarfish hydrolysate was rich in <1,000 Da
(92%) oligopeptides (di- to penta-peptides) (Parthsarathy
et al. 2019). DPP-IV inhibition activities of tuna cooking
juice protease XXIII hydrolysate-extracted oligopeptides,
PGVGGPLGPIGPCYE and CAYQWQRPVDRIR, were
enhanced up to 12% post-GI digestion yet the efficacy of
orientase hydrolysate-isolated PACGGFYISGRPG was not
different pre- and post-digestion (S.-L. Huang et al. 2012).
Immunoreactive calcitonin gene-related peptide (im-CGRP)
like molecules of siki hydrolysate were stable to digestion
and proteases but not the CGRP-like molecules with specific
CGRP binding sites interaction capacity (Martínez-Alvarez
etal. 2012). CGRP neuropeptide molecules have a wide range
of bioactivities of which some also surpass the potencies of
conventional drugs. Among many bioactivities these com-
pounds have great immunomodulating, vasodilating and
satiety effects (Pilon et al. 2011). Levels of CGRP are fairly
low in diabetic and coronary artery patients (L. Wang et al.
2012). Thus, pharmacological properties of these peptides
Table 7. Drug-likeness and ADME analysis of selected sh anti-MetS oligopeptides by Lipinskis rules.
Lipinski’s rule of ve
Molecular
weight (g mol−1)
Lipophilicity
(MLog P)
Hydrogen bond
acceptors
Hydrogen bond
donors
No. of rule
violations Drug-likeness
Sequence
Less than 500
Dalton Less than 5 Less than 5 Less than 10
Less than two
violations
Follows
Lipinski’s rule
Bioavailability
score*
DP 230.22 −1.27 641Ye s 0.56
FF 312.36 1.9 430Ye s 0.55
I/LD 246.26 −0.58 641Yes 0.56
I/LH 268.31 −0.94 541Yes 0.55
PP 212.25 −0.07 420Yes 0.55
VC 220.29 −0.3 430Yes 0.55
VP 214.26 −0.07 430Ye s 0.55
VW 303.36 0.56 440Yes 0.55
YP 278.3 0.22 541Yes 0.55
FSD 367.35 −1.23 861Yes 0.11
FVA 335.4 0.61 541Ye s 0.55
GPA 243.26 −1.38 531Ye s 0.55
GPL 285.34 −0.53 531Yes 0.55
IPI 341.45 0.48 531Yes 0.55
WSG 348.35 −1.48 661Yes 0.55
APA G 314.34 −1.78 661Yes 0.55
GGYE 424.41 −1.67 971Ye s 0.11
GPAE 372.37 −2.07 851Yes 0.11
GPAV, LGPG 342.39 −1.26 641Ye s 0.55
GPGA 300.31 −2.06 641Yes 0.55
PGPL 382.45 −0.53 631Yes 0.55
YYPL 554.63 0.33 862No 0.17
GEYGP 521.52 −2.11 10 8 2 No 0.11
IIAEK 572.69 −1.13 10 8 2 No 0.17
Drug likeness of selected di-, tri-, tetra- and penta-peptides were predicted according to Lipinski’s rule of ve using SwissADME web tool. *Abbot bioavailability
score: probability of a compound to have at least 10% oral bioavailability in rat.
40 S. ABACHI ETAL.
are possibly ideal for the research and development of
anti-MetS drugs and selected ones have been analyzed for
their ADME properties (Table 7).
Conclusion and possible future directions
Fish biopeptides derived by a wide range of techniques,
mostly enzymatic hydrolysis, membrane filtration as well as
chromatographic ones, have shown to prevent and alleviate
MetS and its main associated risk factors in great number
of studies. About half of the studies made efficient use of
fish byproducts and processing leftovers. Nevertheless, more
attention shall be given to the problem of food security and
environmental protection act including greenhouse gas emis-
sion thus exploiting food byproducts and underutilized spe-
cies of fish in as many studies as possible and contributing
to the related global strategies. It is noteworthy that many
of the peptides reviewed herein are of multifunctional nature
thus of high importance to the researchers and product
developers whether in pharmaceutical, nutraceutical and/or
food industry. Mortality and morbidity rates of metabolic
diseases are significant and the prevention and/or the ther-
apy options are limited due to several reasons such as non-
compliance issue and side effects making the natural
non-cytotoxic therapeutic treatments important in this era.
Hypoglycemic, hypolipidemic and anti-obesogenic fish bio-
active peptides exert their effect via different mechanisms
of which many are yet not well studied and understood.
These biomolecules could be involved as drug leads in drug
discoveries and/or simply as active ingredients in nutraceu-
ticals applicable to clinical nutrition and/or to functional
food. Bioavailability of the fish peptides was high in many
of the studies we have reviewed and discussed in this article
since integrity of the biomolecules was not negatively
affected and/or hindered by the digestive enzymes in various
animal models and human subjects. Yet, for a definite con-
clusion full capacity of fish anti-MetS peptides must be
tested in human subjects including ill and healthy ones.
Disclosure statement
No potential conict of interest was reported by the authors.
Funding
is project was made possible by Natural Sciences and Engineering
Research Council of Canada (NSERC) under the name of Strategic
Partnership Grants for Projects grant STPGP/479527-2015.
ORCID
Lucie Beaulieu http://orcid.org/0000-0001-8120-1039
References
Aadland, E. K., I. E. Gra, C. Lavigne, Ø. Eng, M. Paquette, A. Holthe,
G. Mellgren, L. Madsen, H. Jacques, and B. Liaset. 2016. Lean
seafood intake reduces postprandial C-peptide and lactate concen-
trations in healthy adults in a randomized controlled trial with a
crossover design. e Journal of Nutrition 146 (5):1027–34. doi:
10.3945/jn.115.229278.
Aadland, E. K., C. Lavigne, I. E. Gra, Ø. Eng, M. Paquette, A. Holthe,
G. Mellgren, H. Jacques, and B. Liaset. 2015. Lean-seafood intake
reduces cardiovascular lipid risk factors in healthy subjects: Results
from a randomized controlled trial with a crossover design, 2. e
American Journal of Clinical Nutrition 102 (3):582–92. doi: 10.3945/
ajcn.115.112086.
Abachi, S., L. Bazinet, and L. Beaulieu. 2019. Antihypertensive and
Angiotensin-I-Converting Enzyme (ACE)-inhibitory peptides from
sh as potential cardioprotective compounds. Marine Drugs 17
(11):613–52. doi: 10.3390/md17110613.
Abachi, S., C. Oret, I. Fliss, A. Marette, L. Bazinet, and L. Beaulieu.
2022. Isolation of immuno-modulatory biopeptides from Atlantic
mackerel (Scomber scombrus) protein hydrolysate based on molec-
ular weight, charge, and hydrophobicity. Food and Bioprocess
Tech nol o g y . doi: 10.1007/s11947-022-02786-4.
Abachi, S., G. Pilon, A. Marette, L. Bazinet, and L. Beaulieu. 2021.
Immunomodulatory eects of sh peptides on cardiometabolic syn-
drome associated risk factors: A review. Food Reviews International.
1–44. doi: 10.1080/87559129.2021.2014861.
Abdelhedi, O., H. Khemakhem, R. Nasri, M. Jridi, L. Mora, I. Ben
Amor, K. Jamoussi, F. Toldrá, J. Gargouri, and M. Nasri. 2019.
Assessment of cholesterol, glycemia control and short-and long-term
antihypertensive effects of smooth hound viscera peptides in
high-salt and fructose diet-fed wistar rats. Marine Drugs 17 (4):194–
213. doi: 10.3390/md17040194.
Abete, I., A. Astrup, J. A. Martínez, I. orsdottir, and M. A. Zulet.
2010. Obesity and the metabolic syndrome: Role of dierent dietary
macronutrient distribution patterns and specic nutritional compo-
nents on weight loss and maintenance. Nutrition Reviews 68 (4):214–
31. doi: 10.1111/j.1753-4887.2010.00280.x.
Aane, F., S. Louala, N. el Imane Harrat, F. Bensalah, H. Chekkal, A.
Allaoui, and M. Lamri-Senhadji. 2018. Hypolipidemic, antioxidant
and antiatherogenic property of sardine by-products proteins in
high-fat diet induced obese rats. Life Sciences 199:16–22. doi:
10.1016/j.lfs.2018.03.001.
Ait-Yahia, D., S. Madani, J.-L. Savelli, J. Prost, M. Bouchenak, and J.
Belleville. 2003. Dietary sh protein lowers blood pressure and
alters tissue polyunsaturated fatty acid composition in spontaneous-
ly hypertensive rats. Nutrition 19 (4):342–6. doi: 10.1016/
S0899-9007(02)00858-4.
Alamshah, A., E. Spreckley, M. Norton, J. S. Kinsey-Jones, A. Amin,
A. Ramgulam, Y. Cao, R. Johnson, K. Saleh, E. Akalestou, et al.
2017. L-phenylalanine modulates gut hormone release and glucose
tolerance, and suppresses food intake through the calcium-sensing
receptor in rodents. International Journal of Obesity (2005) 41
(11):1693–701. doi: 10.1038/ijo.2017.164.
Ali, M. Y., H. J. Jung, S. Jannat, H. A. Jung, and J. S. Choi. 2016. In
vitro antidiabetic and antioxidant potential of the ethanolic extract
of skipjack tuna (K atsuwonus Pelamis) heart. Journal of Food
Biochemistry 40 (4):583–92. doi: 10.1111/jc.12257.
Alvarado-Vásquez, N., R. Lascurain, E. Cerón, B. Vanda, G.
Carvajal-Sandoval, A. Tapia, J. Guevara, L. F. Montaño, and E.
Zenteno. 2006. Oral glycine administration attenuates diabetic com-
plications in streptozotocin-induced diabetic rats. Life Sciences 79
(3):225–32. doi: 10.1016/j.lfs.2005.12.055.
Alvarado-Vásquez, N., P. Zamudio, E. Cerón, B. Vanda, E. Zenteno,
and G. Carvajal-Sandoval. 2003. Effect of glycine in
streptozotocin-induced diabetic rats. Comparative Biochemistry and
Physiology Part C: Toxicology & Pharmacology 134 (4):521–7.
Arakawa, M., T. Masaki, J. Nishimura, M. Seike, and H. Yoshimatsu.
2011. e eects of branched-chain amino acid granules on the
accumulation of tissue triglycerides and uncoupling proteins in
diet-induced obese mice. Endocrine Journal 58 (3):161–70. doi:
10.1507/endocrj.K10E-221.
Araujo, T. R., I. N. Freitas, J. F. Vettorazzi, T. M. Batista, J. C.
Santos-Silva, M. L. Boneur, S. L. Balbo, A. C. Boschero, E. M.
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 41
Carneiro, and R. A. Ribeiro. 2017. Benets of L-alanine or L-arginine
supplementation against adiposity and glucose intolerance in mono-
sodium glutamate-induced obesity. European Journal of Nutrition
56 (6):2069–80. doi: 10.1007/s00394-016-1245-6.
Astre, G., S. Deleruyelle, A. Dortignac, C. Bonnet, P. Valet, and C.
Dray. 2018. Diet-induced obesity and associated disorders are pre-
vented by natural bioactive type 1 sh collagen peptides (Naticol®)
treatment. Journal of Physiology and Biochemistry 74 (4):647–54.
doi: 10.1007/s13105-018-0650-0.
Athmani, N., F. Dehiba, A. Allaoui, A. Barkia, A. Bougatef, M. Y.
Lamri-Senhadji, M. Nasri, and A. Boualga. 2015. Sardina pilchardus
and Sardinella aurita protein hydrolysates reduce cholesterolemia and
oxidative stress in rat fed high cholesterol diet. Journal of Experimental
and Integrative Medicine 5 (1):47–54. doi: 10.5455/jeim.180115.or.119.
Ayabe, T., T. Mizushige, W. Ota, F. Kawabata, K. Hayamizu, L. Han,
T. Tsuji, R. Kanamoto, and K. Ohinata. 2015. A novel Alaska
pollack-derived peptide, which increases glucose uptake in skeletal
muscle cells, lowers the blood glucose level in diabetic mice. Food
& Function 6 (8):2749–57. doi: 10.1039/c5fo00401b.
Ayaso, R., H. Ghattas, M. Abiad, and O. Obeid. 2014. Meal pattern
of male rats maintained on amino acid supplemented diets: e
eect of tryptophan, lysine, arginine, proline and threonine. Nutrients
6 (7):2509–22. doi: 10.3390/nu6072509.
Bassil, M. S., N. Hwalla, and O. A. Obeid. 2007. Meal pattern of male
rats maintained on histidine‐, leucine‐, or tyrosine‐supplemented
diet. Obesity 15 (3):616–23. doi: 10.1038/oby.2007.565.
Bauvois, B. 1988. A collagen-binding glycoprotein on the surface of
mouse broblasts is identied as dipeptidyl peptidase IV. e
Biochemical Journal 252 (3):723–31. doi: 10.1042/bj2520723.
Benomar, S., S. Yahia, F. Dehiba, N. Guillen, M. J. Rodriguez-Yoldi,
J. Osada, and A. Boualga. 2015. Dierential antioxidative and hy-
pocholesterolemic responses to two sh protein hydrolysates (Sardina
pilchardus and Boops boops) in cholesterol-fed rats. Nutrition &
Food Science 45 (3):448–66. doi: 10.1108/NFS-11-2014-0096.
Bernstein, A. M., Q. Sun, F. B. Hu, M. J. Stampfer, J. E. Manson, and
W. C. Willett. 2010. Major dietary protein sources and risk of cor-
onary heart disease in women. Circulation 122 (9):876–83. doi:
10.1161/CIRCULATIONAHA.109.915165.
Binder, E., F. J. Bermúdez-Silva, C. André, M. Elie, S. Y. Romero-Zerbo,
T. Leste-Lasserre, l. Belluomo, A. Duchampt, S. Clark, A. Aubert,
etal. 2013. Leucine supplementation protects from insulin resistance
by regulating adiposity levels. PLoS One. 8 (9):e74705–12. doi:
10.1371/journal.pone.0074705.
Binder, E., F. J. Bermúdez-Silva, M. Elie, T. Leste-Lasserre, I. Belluomo,
S. Clark, A. Duchampt, G. Mithieux, and D. Cota. 2014. Leucine
supplementation modulates fuel substrates utilization and glucose
metabolism in previously obese mice. Obesity (Silver Spring, MD)
22 (3):713–20. doi: 10.1002/oby.20578.
Bjørndal, B., C. Berge, M. S. Ramsvik, A. Svardal, P. Bohov, J. Skorve,
and R. K. Berge. 2013. A sh protein hydrolysate alters fatty acid
composition in liver and adipose tissue and increases plasma car-
nitine levels in a mouse model of chronic inammation. Lipids in
Health and Disease 12 (1):1–11. doi: 10.1186/1476-511X-12-143.
Blümer, R. M., C. P. van Roomen, A. J. Meijer, J. H. Houben-Weerts,
H. P. Sauerwein, and P. F. Dubbelhuis. 2008. Regulation of adi-
ponectin secretion by insulin and amino acids in 3T3-L1 adipocytes.
Metabolism: clinical and Experimental 57 (12):1655–62. doi: 10.1016/j.
metabol.2008.07.020.
Børsheim, E., Q.-U. T. Bui, S. Tissier, M. G. Cree, O. Rønsen, B. Morio,
A. A. Ferrando, H. Kobayashi, B. R. Newcomer, and R. R. Wolfe.
2009. Amino acid supplementation decreases plasma and liver tri-
acylglycerols in elderly. Nutrition 25 (3):281–8. doi: 10.1016/j.
nut.2008.09.001.
Borzoei, S., M. Neovius, B. Barkeling, A. Teixeira-Pinto, and S. Rössner.
2006. A comparison of eects of sh and beef protein on satiety
in normal weight men. European Journal of Clinical Nutrition 60
(7):897–902. doi: 10.1038/sj.ejcn.1602397.
Bougatef, A., R. Ravallec, N. Nedjar-Arroume, A. Barkia, D. Guillochon,
and M. Nasri. 2010. Evidence of in vivo satietogen eect and con-
trol of food intake of smooth hound (Mustelus mustelus) muscle
protein hydrolysate in rats. Journal of Functional Foods 2 (1):10–6.
doi: 10.1016/j.j.2009.11.001.
Burke, A. P., F. D. Kolodgie, A. Zieske, D. R. Fowler, D. K. Weber, P.
J. Varghese, A. Farb, and R. Virmani. 2004. Morphologic ndings
of coronary atherosclerotic plaques in diabetics: A postmortem
study. Arteriosclerosis, rombosis, and Vascular Biology 24 (7):1266–
71. doi: 10.1161/01.ATV.0000131783.74034.97.
Cai, Z., W. Li, K. Mai, W. Xu, Y. Zhang, and Q. Ai. 2015. Eects of
dietary size-fractionated sh hydrolysates on growth, activities of
digestive enzymes and aminotransferases and expression of some
protein metabolism related genes in large yellow croaker
(Larimichthys crocea) larvae. Aquaculture 440:40–7. doi: 10.1016/j.
aquaculture.2015.01.026.
Caldow, M. K., D. J. Ham, D. P. Godeassi, A. Chee, G. S. Lynch, and
R. Koopman. 2016. Glycine supplementation during calorie restric-
tion accelerates fat loss and protects against further muscle loss in
obese mice. Clinical Nutrition 35 (5):1118–26. doi: 10.1016/j.
clnu.2015.08.013.
Carneiro, E. M., M. Q. Latorraca, E. Araujo, M. Beltrá, M. J. Oliveras,
M. Navarro, G. Berná, F. J. Bedoya, L. A. Velloso, B. Soria, et al.
2009. Taurine supplementation modulates glucose homeostasis and
islet function. e Journal of Nutritional Biochemistry 20 (7):503–11.
doi: 10.1016/j.jnutbio.2008.05.008.
Chan, D. C., H. P. Barrett, and G. F. Watts. 2004. Dyslipidemia in
visceral obesity: mechanisms, implications, and therapy. American
Journal of Cardiovascular Drugs: Drugs, Devices, and Other
Interventions 4 (4):227–46. doi: 10.2165/00129784-200404040-00004.
Chen, H., D. Simar, J. Ting, J. Erkelens, and M. Morris. 2012. Leucine
improves glucose and lipid status in ospring from obese dams,
dependent on diet type, but not caloric intake. Journal of
Neuroendocrinology 24 (10):1356–64. doi:
10.1111/j.1365-2826.2012.02339.x.
Cheung, I. W, and E. C. Li-Chan. 2017. Enzymatic production of
protein hydrolysates from steelhead (Oncorhynchus mykiss) skin
gelatin as inhibitors of dipeptidyl-peptidase IV and angiotensin-I
converting enzyme. Journal of Functional Foods 28:254–64. doi:
10.1016/j.j.2016.10.030.
Chevrier, G., P. L. Mitchell, L.-E. Rioux, F. Hasan, T. Jin, C. R. Roblet,
A. Doyen, G. Pilon, P. St-Pierre, C. Lavigne, et al. 2015.
Low-molecular-weight peptides from salmon protein prevent
obesity-linked glucose intolerance, inammation, and dyslipidemia
in LDLR−/−/ApoB100/100 mice–3. e Journal of Nutrition 145
(7):1415–22. doi: 10.3945/jn.114.208215.
Choi, M.-J., J.-H. Kim, and K. J. Chang. 2006. e eect of dietary
taurine supplementation on plasma and liver lipid concentrations
and free amino acid concentrations in rats fed a high-cholesterol
diet. Taurine 6:235–42.
Choi, K.-W., H.-J. Park, D.-H. Jung, T.-W. Kim, Y.-M. Park, B.-O. Kim,
E.-H. Sohn, E.-Y. Moon, S. H. Um, D.-K. Rhee, et al. 2010.
Inhibition of TNF-α-induced adhesion molecule expression by di-
osgenin in mouse vascular smooth muscle cells via downregulation
of the MAPK, Akt and NF-κB signaling pathways. Vascular
Pharmacology 53 (5–6):273–80. doi: 10.1016/j.vph.2010.09.007.
Cicero, A. F, and A. Colletti. 2016. Role of phytochemicals in the
management of metabolic syndrome. Phytomedicine: International
Journal of Phytotherapy and Phytopharmacology 23 (11):1134–44.
doi: 10.1016/j.phymed.2015.11.009.
Cojocaru, E., N. Filip, C. Ungureanu, C. Filip, and M. Danciu. 2014.
Eects of valine and leucine on some antioxidant enzymes in hy-
percholesterolemic rats. Health 6 (17):2313–21. doi: 10.4236/
health.2014.617266.
Combs, T. P., A. H. Berg, S. Obici, P. E. Scherer, and L. Rossetti. 2001.
Endogenous glucose production is inhibited by the adipose-derived
protein Acrp30. e Journal of Clinical Investigation 108 (12):1875–
81. doi: 10.1172/JCI14120.
Cudennec, B., M. Fouchereau-Peron, F. Ferry, E. Duclos, and R.
Ravallec. 2012. In vitro and in vivo evidence for a satiating eect
of sh protein hydrolysate obtained from blue whiting (Micromesistius
poutassou) muscle. Journal of Functional Foods 4 (1):271–7. doi:
10.1016/j.j.2011.12.003.
42 S. ABACHI ETAL.
Cudennec, B., R. Ravallec-Plé, E. Courois, and M. Fouchereau-Peron.
2008. Peptides from sh and crustacean by-products hydrolysates
stimulate cholecystokinin release in STC-1 cells. Food Chemistry
111 (4):970–5. doi: 10.1016/j.foodchem.2008.05.016.
Cui-Feng, Z., L. Guan-Zhi, P. Hong-Bin, F. Zhang, C. Yun, and L.
Yong. 2010. Eect of marine collagen peptides on markers of met-
abolic nuclear receptors in type 2 diabetic patients with/without
hypertension. Biomedical and Environmental Sciences: BES 23
(2):113–20. doi: 10.1016/S0895-3988(10)60040-2.
Dale, H. F., C. Jensen, T. Hausken, E. Lied, J. G. Hatlebakk, I. Brønstad,
D. A. L. Ho, and G. A. Lied. 2018. Eect of a cod protein hy-
drolysate on postprandial glucose metabolism in healthy subjects:
A double-blind cross-over trial. Journal of Nutritional Science 7:1–
9. doi: 10.1017/jns.2018.23.
de Mello, V. D. F., A. T. Erkkilä, U. S. Schwab, L. Pulkkinen, M.
Kolehmainen, M. Atalay, H. Mussalo, M. Lankinen, M. Orešič, S.
Lehto, etal. 2009. e eect of fatty or lean sh intake on inam-
matory gene expression in peripheral blood mononuclear cells of
patients with coronary heart disease. European Journal of Nutrition
48 (8):447–55. doi: 10.1007/s00394-009-0033-y.
Delzenne, N. M, and P. D. Cani. 2005. A place for dietary bre in
the management of the metabolic syndrome. Current Opinion in
Clinical Nutrition and Metabolic Care 8 (6):636–40. doi: 10.1097/01.
mco.0000171124.06408.71.
Demonty, I., Y. Deshaies, and H. Jacques. 1998. Dietary proteins mod-
ulate the eects of sh oil on triglyceridemia in the rat. Lipids 33
(9):913–21. doi: 10.1007/s11745-998-0288-5.
Diakogiannaki, E., R. Pais, G. Tolhurst, H. E. Parker, J. Horscro, B.
Rauscher, T. Zietek, H. Daniel, F. M. Gribble, and F. Reimann. 2013.
Oligopeptides stimulate glucagon-like peptide-1 secretion in mice
through proton-coupled uptake and the calcium-sensing receptor.
Diabetologia 56 (12):2688–96. doi: 10.1007/s00125-013-3037-3.
Dillon, E. L., M. Sheeld-Moore, D. Paddon-Jones, C. Gilkison, A. P.
Sanford, S. L. Casperson, J. Jiang, D. L. Chinkes, and R. J. Urban.
2009. Amino acid supplementation increases lean body mass, bas-
al muscle protein synthesis, and insulin-like growth factor-I expres-
sion in older women. e Journal of Clinical Endocrinology &
Metabolism 94 (5):1630–7. doi: 10.1210/jc.2008-1564.
Dort, J., A. Sirois, N. Leblanc, C. H. Côté, and H. Jacques. 2012.
Benecial eects of cod protein on skeletal muscle repair following
injury. Applied Physiology, Nutrition, and Metabolism = Physiologie
Appliquee, Nutrition et Metabolisme 37 (3):489–98. doi: 10.1139/
h2012-021.
Drotningsvik, A., S. A. Mjøs, I. Høgøy, T. Remman, and O. A.
Gudbrandsen. 2015. A low dietary intake of cod protein is sucient
to increase growth, improve serum and tissue fatty acid composi-
tions, and lower serum postprandial glucose and fasting non-esteried
fatty acid concentrations in obese Zucker fa/fa rats. European Journal
of Nutrition 54 (7):1151–60. doi: 10.1007/s00394-014-0793-x.
Drotningsvik, A., S. A. Mjøs, D. M. Pampanin, R. Slizyte, A. Carvajal,
T. Remman, I. Høgøy, and O. A. Gudbrandsen. 2016. Dietary sh
protein hydrolysates containing bioactive motifs aect serum and
adipose tissue fatty acid compositions, serum lipids, postprandial
glucose regulation and growth in obese Zucker fa/fa rats. e British
Journal of Nutrition 116 (8):1336–45. doi: 10.1017/S0007114516003548.
Drotningsvik, A., D. M. Pampanin, R. Slizyte, A. Carvajal, I. Høgøy,
T. Remman, and O. A. Gudbrandsen. 2018. Hydrolyzed proteins
from herring and salmon rest raw material contain peptide motifs
with angiotensin-I converting enzyme inhibitors and resulted in
lower urine concentrations of protein, cystatin C and glucose when
fed to obese Zucker fa/fa rats. Nutrition Research (New York, NY)
52:14–21. doi: 10.1016/j.nutres.2018.01.001.
Drotningsvik, A., L. Vikøren, S. Mjøs, Å. Oterhals, D. Pampanin, O.
Flesland, and O. Gudbrandsen. 2018. Water-soluble sh protein
intake led to lower serum and liver cholesterol concentrations in
obese zucker fa/fa rats. Marine Drugs 16 (5):149. doi: 10.3390/
md16050149.
Durand, R., E. Fraboulet, A. Marette, and L. Bazinet. 2019. Simultaneous
double cationic and anionic molecule separation from herring milt
hydrolysate and impact on resulting fraction bioactivities. Separation
and Purification Technology 210:431–41. doi: 10.1016/j.sep-
pur.2018.08.017.
Elvevoll, E. O., K.-E. Eilertsen, J. Brox, B. T. Dragnes, P. Falkenberg,
J. O. Olsen, B. Kirkhus, A. Lamglait, and B. Østerud. 2008. Seafood
diets: Hypolipidemic and antiatherogenic eects of taurine and n-3
fatty acids. Atherosclerosis 200 (2):396–402. doi: 10.1016/j.athero-
sclerosis.2007.12.021.
Emery, C., E. Torreton, S. Dejager, L. Levy-Bachelot, S. Bineau, and
B. Detournay. 2020. Cost of managing type 2 diabetes before and
aer initiating dipeptidyl peptidase 4 inhibitor treatment: A longi-
tudinal study using a French public health insurance database.
Diabetes erapy 11 (2):535–14. doi: 10.1007/s13300-020-00760-x.
Erkkilä, A. T., U. S. Schwab, S. Lehto, V. D. de Mello, A. J. Kangas,
P. Soininen, M. Ala-Korpela, and M. I. Uusitupa. 2014. Eect of
fatty and lean sh intake on lipoprotein subclasses in subjects with
coronary heart disease: A controlled trial. Journal of Clinical
Lipidology 8 (1):126–33. doi: 10.1016/j.jacl.2013.09.007.
Espe, M., R. M. Rathore, Z.-Y. Du, B. Liaset, and A. El-Mowa. 2010.
Methionine limitation results in increased hepatic FAS activity, high-
er liver 18:1 to 18:0 fatty acid ratio and hepatic TAG accumulation
in Atlantic salmon, Salmo salar. Amino Acids 39 (2):449–60. doi:
10.1007/s00726-009-0461-2.
Feigh, M., K. V. Andreassen, A. V. Neutzsky-Wul, S. T. Petersen, C.
Hansen, A. C. Bay-Jensen, J. E. Henriksen, H. Beck-Nielsen, C.
Christiansen, K. Henriksen, et al. 2012. Oral salmon calcitonin
attenuates hyperglycaemia and preserves pancreatic beta-cell area
and function in Zucker diabetic fatty rats . British Journal of
Pharmacology 167 (1):151–63. doi: 10.1111/j.1476-5381.2012.01979.x.
Feigh, M., K. Henriksen, K. Andreassen, C. Hansen, J. Henriksen, H.
Beck‐Nielsen, C. Christiansen, and M. Karsdal. 2011. A novel oral
form of salmon calcitonin improves glucose homeostasis and re-
duces body weight in diet‐induced obese rats. Diabetes, Obesity
and Metabolism 13 (10):911–20. doi:
10.1111/j.1463-1326.2011.01425.x.
Feng, R., Y. Niu, X. Sun, Q. Li, C. Zhao, C. Wang, F. Guo, C. Sun,
and Y. Li. 2013. Histidine supplementation improves insulin resis-
tance through suppressed inammation in obese women with the
metabolic syndrome: A randomised controlled trial. Diabetologia 56
(5):985–94. doi: 10.1007/s00125-013-2839-7.
Freudenberg, A., K. J. Petzke, and S. Klaus. 2013. Dietary L-leucine
and L-alanine supplementation have similar acute eects in the
prevention of high-fat diet-induced obesity. Amino Acids 44 (2):519–
28. doi: 10.1007/s00726-012-1363-2.
Geraedts, M. C., F. J. Troost, M. A. Fischer, L. Edens, and W. H. Saris.
2011. Direct induction of CCK and GLP-1 release from murine
endocrine cells by intact dietary proteins. Molecular Nutrition &
Food Research 55 (3):476–84. doi: 10.1002/mnfr.201000142.
Greeneld, J. R., I. S. Farooqi, J. M. Keogh, E. Henning, A. M. Habib,
A. Blackwood, F. Reimann, J. J. Holst, and F. M. Gribble. 2009.
Oral glutamine increases circulating glucagon-like peptide 1, glu-
cagon, and insulin concentrations in lean, obese, and type 2 dia-
betic subjects. e American Journal of Clinical Nutrition 89 (1):106–
13. doi: 10.3945/ajcn.2008.26362.
Grieger, J. A., M. D. Miller, and L. Cobiac. 2014. Investigation of the
eects of a high sh diet on inammatory cytokines, blood pressure,
and lipids in healthy older Australians. Food & Nutrition Research
58 (1):20369–11. doi: 10.3402/fnr.v58.20369.
Grintescu, I. M., I. L. Vasiliu, I. C. Badica, L. Mirea, D. Pavelescu, A.
Balanescu, and I. C. Grintescu. 2015. e inuence of parenteral
glutamine supplementation on glucose homeostasis in critically ill
polytrauma patients-A randomized-controlled clinical study . Clinical
Nutrition (Edinburgh, Scotland) 34 (3):377–82. doi: 10.1016/j.
clnu.2014.05.006.
Gudbrandsen, O. A., H. Wergedahl, B. Liaset, M. Espe, and R. K.
Berge. 2005. Dietary proteins with high isoavone content or low
methionine-glycine and lysine-arginine ratios are hypocholestero-
laemic and lower the plasma homocysteine level in male Zucker
fa/fa rats. e British Journal of Nutrition 94 (3):321–30. doi:
10.1079/bjn20051496.
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 43
Guenther, M., R. James, J. Marks, S. Zhao, A. Szabo, and S. Kidambi.
2014. Adiposity distribution inuences circulating adiponectin lev-
els. Translational Research 164 (4):270–7. doi: 10.1016/j.
trsl.2014.04.008.
Guérard, F., N. Decourcelle, C. Sabourin, C. Floch-Laizet, L. Le Grel,
P. Le Floc’H, F. Gourlay, R. Le Delezir, P. Jaouen, and P. Bourseau.
2010. Recent developments of marine ingredients for food and
nutraceutical applications: A review. Journal Des Sciences Halieutique
et Aquatique 2:21–7.
Gunnarsdottir, I., H. Tomasson, M. Kiely, J. Martinez, N. Bandarra,
M. Morais, and I. orsdottir. 2008. Inclusion of sh or sh oil in
weight-loss diets for young adults: Effects on blood lipids.
International Journal of Obesity (2005) 32 (7):1105–13. doi: 10.1038/
ijo.2008.64.
Gunnerud, U. J., C. Heinzle, J. J. Holst, E. M. Östman, and I. M.
Björck. 2012. Eects of pre-meal drinks with protein and amino
acids on glycemic and metabolic responses at a subsequent com-
posite meal. PLoS One. 7 (9):e44731–9. doi: 10.1371/journal.
pone.0044731.
Guo, K., Y.-H. Yu, J. Hou, and Y. Zhang. 2010. Chronic leucine sup-
plementation improves glycemic control in etiologically distinct
mouse models of obesity and diabetes mellitus. Nutrition &
Metabolism 7 (1):57–67. doi: 10.1186/1743-7075-7-57.
Hamza‐Reguig, S., S. Louala, A. Boualga, and M. Y. Lamri-Senhadji.
2013. Eect of sardine protein on redox status in hypercholester-
olemic rats. Nutrition & Food Science 43 (3):277–84. doi:
10.1108/00346651311327936.
Harnedy, P. A., V. Parthsarathy, C. M. McLaughlin, M. B. O’Keee, P.
J. Allsopp, E. M. McSorley, F. P. O’Harte, and R. J. FitzGerald.
2018a. Atlantic salmon (Salmo salar) co-product-derived protein
hydrolysates: A source of antidiabetic peptides. Food Research
International 106:598–606. doi: 10.1016/j.foodres.2018.01.025.
Harnedy, P. A., V. Parthsarathy, C. M. McLaughlin, M. B. O’Keee, P.
J. Allsopp, E. M. McSorley, F. P. O’Harte, and R. J. FitzGerald.
2018b. Blue whiting (Micromesistius poutassou) muscle protein
hydrolysate with in vitro and in vivo antidiabetic properties. Journal
of Functional Foods 40:137–45. doi: 10.1016/j.j.2017.10.045.
Hatanaka, T., K. Kawakami, and M. Uraji. 2014. Inhibitory eect of
collagen-derived tripeptides on dipeptidylpeptidase-IV activity.
Journal of Enzyme Inhibition and Medicinal Chemistry 29 (6):823–8.
doi: 10.3109/14756366.2013.858143.
Henaux, L., J. ibodeau, G. Pilon, T. Gill, A. Marette, and L. Bazinet.
2019. How charge and triple size-selective membrane separation of
peptides from salmon protein hydrolysate orientate their biological
response on glucose uptake. International Journal of Molecular
Sciences 20 (8):1939–54. doi: 10.3390/ijms20081939.
Heraief, E., P. Burckhardt, J. J. Wurtman, and R. J. Wurtman. 1985.
Tryptophan administration may enhance weight loss by some mod-
erately obese patients on a protein‐sparing modied fast (PSMF)
diet. International Journal of Eating Disorders 4 (3):281–92. doi:
10.1002/1098-108X(198508)4:3<281::AID-EAT22600403
05>3.0.CO;2-6.
Hirche, F., A. Schröder, B. Knoth, G. Stangl, and K. Eder. 2006. Eect
of dietary methionine on plasma and liver cholesterol concentrations
in rats and expression of hepatic genes involved in cholesterol
metabolism. British Journal of Nutrition 95 (5):879–88. doi: 10.1079/
BJN20061729.
Hokmabadinazhad, S. A., J.-P. Songpadith, V. P. Houde, G. Pilon, I.
Fliss, A. Marette, L. Bazinet, and L. Beaulieu. 2022. Bioactivity of
mackerel peptides on obesity and insulin resistance, an in-vivo
study. Food Bioscience 47:101641. doi: 10.1016/j.io.2022.101641.
Holm, L. J., M. Haupt-Jorgensen, J. Larsen, J. D. Giacobini, M. Bilgin,
and K. Buschard. 2018. L-serine supplementation lowers diabetes
incidence and improves blood glucose homeostasis in NOD mice.
Plos One 13 (3):e0194414–11. doi: 10.1371/journal.pone.0194414.
Hosomi, R., M. Fukao, K. Fukunaga, M. Okuno, R. Yagita, S. Kanda,
T. Nishiyama, and M. Yoshida. 2010. Eect of sh protein and
peptides on lipid absorption in rats. Trace Nutr Res 27:21–7.
Hosomi, R., K. Fukunaga, H. Arai, S. Kanda, T. Nishiyama, and M.
Yoshida. 2011. Fish protein decreases serum cholesterol in rats by
inhibition of cholesterol and bile acid absorption. Journal of Food
Science 76 (4):H116–H121. doi: 10.1111/j.1750-3841.2011.02130.x.
Hosomi, R., K. Fukunaga, H. Arai, S. Kanda, T. Nishiyama, and M.
Yoshida. 2012. Fish protein hydrolysates aect cholesterol metabo-
lism in rats fed non-cholesterol and high-cholesterol diets. Journal
of Medicinal Food 15 (3):299–306. doi: 10.1089/jmf.2011.1620.
Hosomi, R., K. Fukunaga, H. Arai, S. Kanda, T. Nishiyama, and M.
Yoshida. 2013. Eect of combination of dietary sh protein and sh
oil on lipid metabolism in rats. Journal of Food Science and
Tech nol o g y 50 (2):266–74. doi: 10.1007/s13197-011-0343-y.
Hosomi, R., K. Fukunaga, H. Arai, T. Nishiyama, and M. Yoshida.
2009. Eects of dietary sh protein on serum and liver lipid con-
centrations in rats and the expression of hepatic genes involved in
lipid metabolism. Journal of Agricultural and Food Chemistry 57
(19):9256–62. doi: 10.1021/jf901954r.
Hosomi, R., H. Maeda, Y. Ikeda, Y. Toda, M. Yoshida, and K. Fukunaga.
2017. Dierential eects of cod proteins and tuna proteins on serum
and liver lipid profiles in rats fed non-cholesterol- and
cholesterol-containing diets. Preventive Nutrition and Food Science
22 (2):90–9. doi: 10.3746/pnf.2017.22.2.90.
Hovland, I. H., I. S. Leikanger, O. Stokkeland, K. H. Waage, S. A.
Mjøs, K. A. Brokstad, A. McCann, P. M. Ueland, R. Slizyte, and A.
Carvajal. 2020. Eects of low doses of sh and milk proteins on
glucose regulation and markers of insulin sensitivity in overweight
adults: A randomised, double blind study. European Journal of
Nutrition 59:1013–1029.
Hsieh, C.-H., T.-Y. Wang, C.-C. Hung, M. Chen, and K.-C. Hsu. 2015.
Improvement of glycemic control in streptozotocin-induced diabet-
ic rats by Atlantic salmon skin gelatin hydrolysate as the
dipeptidyl-peptidase IV inhibitor. Food & Function 6 (6):1887–92.
doi: 10.1039/c5fo00124b.
Hsu, K.-C., Y.-S. Tung, S.-L. Huang, and C.-L. Jao. 2013. Dipeptidyl
peptidase-IV inhibitory activity of peptides in porcine skin gel-
atin hydrolysates. In Bioactive Food Peptides in Health and Disease,
eds. B. Hernandez-Ledesma and C.-C. Hsieh, 205–18. Croatia:
Intech.
Huang, S.-L., C.-L. Jao, K.-P. Ho, and K.-C. Hsu. 2012.
Dipeptidyl-peptidase IV inhibitory activity of peptides derived from
tuna cooking juice hydrolysates. Peptides 35 (1):114–21. doi:
10.1016/j.peptides.2012.03.006.
Huang, F, and W. Wu. 2005. Antidiabetic eect of a new peptide from
Squalus mitsukurii liver (S-8300) in alloxan-diabetes. Clinical and
Experimental Pharmacology & Physiology 32 (7):521–5. doi: 10.1111/j.
1440-1681.2005.04224.x.
Huang, F, and W. Wu. 2010. Study on the antidiabetic mechanism of
a shark liver peptide, S‐8300, in alloxan‐induced diabetes. Journal
of Pharmacy and Pharmacology 61 (6):789–94. doi: 10.1211/
jpp.61.06.0012.
Huang, F. J, and W. T. Wu. 2010. Purication and characterization of
a new peptide (s‐8300) from shark liver. Journal of Food Biochemistry
34 (5):962–70. doi: 10.1111/j.1745-4514.2010.00336.x.
Iba, Y., K. Yokoi, I. Eitoku, M. Goto, S. Koizumi, F. Sugihara, H.
Oyama, and T. Yoshimoto. 2016. Oral administration of collagen
hydrolysates improves glucose tolerance in normal mice through
GLP-1-dependent and GLP-1-independent mechanisms. Journal of
Medicinal Food 19 (9):836–43. doi: 10.1089/jmf.2016.3711.
Inubushi, T., N. Kamemura, M. Oda, J. SAkURAI, Y. NAkAYA, N.
Harada, M. Suenaga, Y. Matsunaga, K. Ishidoh, and N. Katunuma.
2012. L-tryptophan suppresses rise in blood glucose and preserves
insulin secretion in type-2 diabetes mellitus rats. Journal of
Nutritional Science and Vitaminology 58 (6):415–22. doi: 10.3177/
jnsv.58.415.
Ito, T., S. W. Schaer, and J. Azuma. 2012. e potential usefulness
of taurine on diabetes mellitus and its complications. Amino Acids
42 (5):1529–39. doi: 10.1007/s00726-011-0883-5.
Itou, K, and Y. Akahane. 2009. Eect of extracts from heshiko, a
fermented mackerel product, on cholesterol metabolism in Wistar
rats. Fisheries Science 75 (1):241–8. doi: 10.1007/s12562-008-0035-3.
Iwami, K., K. Sakakibara, and F. Ibuki. 1986. Involvement of
post-digestion’hydrophobia’peptides in plasma cholesterol-lowering
44 S. ABACHI ETAL.
eect of dietary plant proteins. Agricultural and Biological Chemistry
50 (5):1217–22.
Iwaniak, A., M. Darewicz, and P. Minkiewicz. 2018. Peptides derived
from foods as supportive diet components in the prevention of
metabolic syndrome. Comprehensive Reviews in Food Science and
Food Safety 17 (1):63–81. doi: 10.1111/1541-4337.12321.
Jain, S. K. 2012. L-cysteine supplementation as an adjuvant therapy
for type-2 diabetes. Canadian Journal of Physiology and Pharmacology
90 (8):1061–4. doi: 10.1139/y2012-087.
Jain, S. K., T. Velusamy, J. L. Croad, J. L. Rains, and R. Bull. 2009.
L-cysteine supplementation lowers blood glucose, glycated hemo-
globin, CRP, MCP-1, and oxidative stress and inhibits NF-κB acti-
vation in the livers of Zucker diabetic rats. Free Radical Biology
and Medicine 46 (12):1633–8. doi: 10.1016/j.freerad-
biomed.2009.03.014.
Javidan, A. N., H. Sabour, M. Nazari, Z. Soltani, R. Heshmat, B.
Larijani, S.-M. Ghodsi, and S.-H E. Razavi. 2017. Is the pattern of
dietary amino acids intake associated with serum lipid prole and
blood pressure among individuals with spinal cord injury? e
Journal of Spinal Cord Medicine 40 (2):201–12. doi:
10.1080/10790268.2015.1109761.
Jemil, I., O. Abdelhedi, R. Nasri, L. Mora, R. Marrekchi, K. Jamoussi,
A. ElFeki, M. Hajji, F. Toldrá, and M. Nasri. 2017. Hypolipidemic,
antiobesity and cardioprotective eects of sardinelle meat our and
its hydrolysates in high-fat and fructose diet fed Wistar rats. Life
Sciences 176:54–66. doi: 10.1016/j.lfs.2016.07.012.
Jemil, I., R. Nasri, O. Abdelhedi, M.-C. Aristoy, R. B. S.-B. Salem,
C. Kallel, R. Marrekchi, K. Jamoussi, A. ElFeki, M. Hajji, et al.
2017. Beneficial effects of fermented sardinelle protein hydrolysates
on hypercaloric diet induced hyperglycemia, oxidative stress and
deterioration of kidney function in wistar rats. Journal of Food
Science and Technology 54 (2):313–25. doi: 10.1007/
s13197-016-2464-9.
Jennings, A., A. MacGregor, T. Pallister, T. Spector, and A. Cassidy.
2016. Associations between branched chain amino acid intake and
biomarkers of adiposity and cardiometabolic health independent of
genetic factors: A twin study. International Journal of Cardiology
223:992–8. doi: 10.1016/j.ijcard.2016.08.307.
Jensen, C., H. F. Dale, T. Hausken, E. Lied, J. G. Hatlebakk, I. Brønstad,
G. A. Lied, and D. A. L. Ho. 2019. Supplementation with cod
protein hydrolysate in older adults: A dose range cross-over study.
Journal of Nutritional Science 8:1–8. doi: 10.1017/jns.2019.37.
Jensen, I.-J., M. Walquist, B. Liaset, E. O. Elvevoll, and K.-E. Eilertsen.
2016. Dietary intake of cod and scallop reduces atherosclerotic
burden in female apolipoprotein E-decient mice fed a Western-type
high fat diet for 13 weeks. Nutrition & Metabolism 13 (8):8–11. doi:
10.1186/s12986-016-0068-z.
Jiang, Y., J. Tang, M. Xie, Z. Wen, S. Qiao, and S. Hou. 2017. reonine
supplementation reduces dietary protein and improves lipid metab-
olism in Pekin ducks. British Poultry Science 58 (6):687–93. doi:
10.1080/00071668.2017.1363871.
Jin, T.Jr, 2013. Separation and purication of antidiabetic bioactive
peptide from salmon and cod waste. Master of Science. Canada:
Dalhousie University.
Jornayvaz, F. R., V. T. Samuel, and G. I. Shulman. 2010. e role of
muscle insulin resistance in the pathogenesis of atherogenic dyslip-
idemia and nonalcoholic fatty liver disease associated with the met-
abolic syndrome. Annual Review of Nutrition 30:273–90. doi:
10.1146/annurev.nutr.012809.104726.
Kasaoka, S., N. Tsuboyama-Kasaoka, Y. Kawahara, S. Inoue, M. Tsuji,
O. Ezaki, H. Kato, T. Tsuchiya, H. Okuda, and S. Nakajima. 2004.
Histidine supplementation suppresses food intake and fat accumu-
lation in rats. Nutrition 20 (11–12):991–6. doi: 10.1016/j.
nut.2004.08.006.
Katan, M. B., L. H. Vroomen, and R. J. Hermus. 1982. Reduction of
casein-induced hypercholesterolaemia and atherosclerosis in rabbits
and rats by dietary glycine, arginine and alanine. Atherosclerosis 43
(2–3):381–91. doi: 10.1016/0021-9150(82)90037-5.
Kato, M., H. Ogawa, T. Kishida, and K. Ebihara. 2011.
Hypocholesterolaemic eect of water-insoluble sh protein from
Alaska pollock in ovariectomised rats is not abolished by methionine
addition. e British Journal of Nutrition 106 (1):57–62. doi: 10.1017/
S0007114510005775.
Kawanishi, N., T. Mizokami, H. Yano, and K. Suzuki. 2013. Exercise
attenuates M1 macrophages and CD8+ T cells in the adipose tissue
of obese mice. Medicine and Science in Sports and Exercise 45
(9):1684–93. doi: 10.1249/MSS.0b013e318289c6.
Kern, M., D. Ellison, Y. Marroquin, M. Ambrose, and K. Mosier. 2002.
Eects of soy protein supplemented with methionine on blood
lipids and adiposity of rats. Nutrition 18 (7–8):654–6. doi: 10.1016/
S0899-9007(02)00783-9.
Ketnawa, S., S. Suwal, J. Y. Huang, and A. M. Liceaga. 2019. Selective
separation and characterisation of dual ACE and DPP‐IV inhibito-
ry peptides from rainbow trout (Oncorhynchus mykiss) protein
hydrolysates. International Journal of Food Science & Technology 54
(4):1062–73. doi: 10.1111/ijfs.13939.
Khaled, H. B., Z. Ghlissi, Y. Chtourou, A. Hakim, N. Ktari, M. A.
Fatma, A. Barkia, Z. Sahnoun, and M. Nasri. 2012. Eect of protein
hydrolysates from sardinelle (Sardinella aurita) on the oxidative
status and blood lipid prole of cholesterol-fed rats. Food Research
International 45 (1):60–8. doi: 10.1016/j.foodres.2011.10.003.
Kim, Y. M., I. H. Kim, J. W. Choi, M. K. Lee, and T. J. Nam. 2015.
e anti-obesity eects of a tuna peptide on 3T3-L1 adipocytes are
mediated by the inhibition of the expression of lipogenic and ad-
ipogenic genes and by the activation of the Wnt/β-catenin signaling
pathway. International Journal of Molecular Medicine 36 (2):327–34.
doi: 10.3892/ijmm.2015.2231.
Kim, Y., M.-J. Kwon, J.-W. Choi, M.-K. Lee, C. Kim, J. Jung, H. Aprianita,
H. Nam, and T.-J. Nam. 2016. Anti-obesity eects of boiled tuna extract
in mice with obesity induced by a high-fat diet. International Journal
of Molecular Medicine 38 (4):1281–8. doi: 10.3892/ijmm.2016.2695.
Kim, S.-J., C. Nian, D. J. Doudet, and C. H. McIntosh. 2009. Dipeptidyl
peptidase IV inhibition with MK0431 improves islet gra survival
in diabetic NOD mice partially via T-cell modulation. Diabetes 58
(3):641–52. doi: 10.2337/db08-1101.
Kim, H.-S., H.-D. Yoon, J.-H. Seong, Y.-G. Lee, C.-L. Xie, S.-H. Kim,
and W.-S. Choi. 2009. Eects of soluble collagen peptides extract
derived from Mugil cephalus scale on the blood glucose and lipid
metabolism in diabetic rats. Journal of Life Science 19 (12):1794–801.
Kittiphattanabawon, P., S. Benjakul, W. Visessanguan, and F. Shahidi.
2013. Inhibition of angiotensin converting enzyme, human LDL
cholesterol and DNA oxidation by hydrolysates from blacktip shark
gelatin. LWT - Food Science and Technology 51 (1):177–82. doi:
10.1016/j.lwt.2012.10.011.
Ko, S.-C, and Y.-J. Jeon. 2013. Marine peptides for preventing meta-
bolic syndrome. Current Protein & Peptide Science 14 (3):183–8.
doi: 10.2174/13892037113149990038.
Konomi, Y., K. Itoh, R. Koga, K. Imai, T. Masuda, S. Abe, M. Tanaka,
and T. Sakata. 2004. Eects of histidine intake on weight loss in
moderately obese women. Nippon Eiyo Shokuryo Gakkaishi 57
(6):265–70. doi: 10.4327/jsnfs.57.265.
Kritchevsky, D., S. A. Tepper, S. K. Czarnecki, and D. M. Klurfeld.
1982. Atherogenicity of animal and vegetable protein. Inuence of
the lysine to arginine ratio. Atherosclerosis 41 (2–3):429–31. doi:
10.1016/0021-9150(82)90208-8.
Ktari, N., O. Belguith-Hadriche, I. Ben Amara, A. Ben Hadj, M. Turki,
F. Makni-Ayedi, T. Boudaouara, A. El Feki, A. Boualga, R. Ben
Salah, et al. 2015. Cholesterol regulatory eects and antioxidant
activities of protein hydrolysates from zebra blenny (Salaria basilis-
ca) in cholesterol-fed rats. Food & Function 6 (7):2273–82. doi:
10.1039/c5fo00492f.
Ktari, N., I. Bkhairia, R. Nasri, R. B. A. Kolsi, R. B. S.-B. Salem, I. B.
Amara, N. Zeghal, B. B. Salah, R. B. Salah, and M. Nasri. 2017.
Zebra blenny protein hydrolysates as a source of bioactive peptides
with prevention eect against oxidative dysfunctions and DNA dam-
age in heart tissues of rats fed a cholesterol-rich diet. Food Research
International (Ottawa, ON) 100 (Pt 1):423–32. doi: 10.1016/j.
foodres.2017.07.040.
Ktari, N., K. Mnafgui, R. Nasri, K. Hamden, I. Bkhairia, A. B. Hadj,
T. Boudaouara, A. Elfeki, and M. Nasri. 2013. Hypoglycemic and
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 45
hypolipidemic eects of protein hydrolysates from zebra blenny
(Salaria basilisca) in alloxan-induced diabetic rats. Food & Function
4 (11):1691–9. doi: 10.1039/c3fo60264h.
Kulakowski, E. C., and J. Maturo. 1984. Hypoglycemic properties of
taurine: Not mediated by enhanced insulin release. Biochemical
Pharmacology 33 (18):2835–8. doi: 10.1016/0006-2952(84)90204-1.
Kumar, L. V., R. J. Shakila, and G. Jeyasekaran. 2019. In vitro
anti-cancer, anti-diabetic, anti-inammation and wound healing
properties of collagen peptides derived from unicorn leatherjacket
(Aluterus Monoceros) at dierent hydrolysis. Turkish Journal of
Fisheries and Aquatic Sciences 19 (7):551–60.
Lancha, A. H., Jr, J. R. Poortmans, and L. O. Pereira. 2009. e eect
of 5 days of aspartate and asparagine supplementation on glucose
transport activity in rat muscle. Cell Biochemistry and Function 27
(8):552–7. doi: 10.1002/cbf.1606.
Larsen, M. O., B. Rolin, U. Ribel, M. Wilken, C. F. Deacon, O.
Svendsen, C. F. Gotfredsen, and R. D. Carr. 2003. Valine pyrrolidide
preserves intact glucose-dependent insulinotropic peptide and im-
proves abnormal glucose tolerance in minipigs with reduced β-cell
mass. Journal of Diabetes Research 4 (2):93–105.
Lassoued, I., M. Mezghani, M. Jridi, F. Rahmouni, K. Jamoussi, T.
Rebai, A. El Feki, M. Nasri, and A. Barkia. 2018. Protective eects
of thornback ray muscle protein hydrolysate against dyslipidemia,
oxidative stress and reduced fertility induced by high cholesterol
diet in adult male rats. RSC Advances 8 (40):22303–12. doi: 10.1039/
C8RA00657A.
Lassoued, I., M. Trigui, Z. Ghlissi, R. Nasri, K. Jamoussi, M. Kessis,
Z. Sahnoun, T. Rebai, A. Boualga, M. Lamri-Senhadji, et al. 2014.
Evaluation of hypocholesterolemic eect and antioxidant activity of
Boops boops proteins in cholesterol-fed rats. Food & Function 5
(6):1224–31. doi: 10.1039/c3fo60705d.
Laviano, A., A. Molno, M. Lacaria, A. Canelli, S. De Leo, I. Preziosa,
and F. R. Fanelli. 2014. Glutamine supplementation favors weight loss
in nondieting obese female patients. A pilot study. European Journal
of Clinical Nutrition 68 (11):1264–6. doi: 10.1038/ejcn.2014.184.
Lavigne, C., A. Marette, and H. Jacques. 2000. Cod and soy proteins
compared with casein improve glucose tolerance and insulin sen-
sitivity in rats. American Journal of Physiology-Endocrinology and
Metabolism 278 (3):E491–E500. doi: 10.1152/ajpendo.2000.278.3.E491.
Lavigne, C., F. Tremblay, G. Asselin, H. Jacques, and A. Marette. 2001.
Prevention of skeletal muscle insulin resistance by dietary cod pro-
tein in high fat-fed rats. American Journal of Physiology. Endocrinology
and Metabolism 281 (1):E62–E71. doi: 10.1152/ajpendo.2001.281.1.E62.
Lee, E. J., J. Hur, S. A. Ham, Y. Jo, S. Lee, M.-J. Choi, and H. G. Seo.
2017. Fish collagen peptide inhibits the adipogenic dierentiation
of preadipocytes and ameliorates obesity in high fat diet-fed mice.
International Journal of Biological Macromolecules 104 (Pt A):281–6.
doi: 10.1016/j.ijbiomac.2017.05.151.
Lee, H., M. Woo, Y. Song, and J. Noh. 2018. Inhibitory eect of skate
skin collagen on hepatic lipid accumulation through regulation of
lipid metabolism. Journal of the Korean Society of Food Science and
Nutrition 47 (3):235–42. doi: 10.3746/jkfn.2018.47.3.235.
Li, Y.-C., Y. Li, L.-Y. Liu, Y. Chen, T.-Q. Zi, S.-S. Du, Y.-S. Jiang,
R.-N. Feng, and C.-H. Sun. 2015. e ratio of dietary branched-chain
amino acids is associated with a lower prevalence of obesity in
young northern Chinese adults: An internet-based cross-sectional
study. Nutrients 7 (11):9573–89. doi: 10.3390/nu7115486.
Liaset, B., L. Madsen, Q. Hao, G. Criales, G. Mellgren, H.-U. Marschall,
P. Hallenborg, M. Espe, L. Frøyland, and K. Kristiansen. 2009. Fish
protein hydrolysate elevates plasma bile acids and reduces visceral
adipose tissue mass in rats. Biochimica et Biophysica Acta 1791
(4):254–62. doi: 10.1016/j.bbalip.2009.01.016.
Liaset, B., J. Øyen, H. Jacques, K. Kristiansen, and L. Madsen. 2019.
Seafood intake and the development of obesity, insulin resistance
and type 2 diabetes. Nutrition Research Reviews 32 (1):146–67. doi:
10.1017/S0954422418000240.
Li-Chan, E. C., S.-L. Hunag, C.-L. Jao, K.-P. Ho, and K.-C. Hsu. 2012.
Peptides derived from Atlantic salmon skin gelatin as
dipeptidyl-peptidase IV inhibitors. Journal of Agricultural and Food
Chemistry 60 (4):973–8. doi: 10.1021/jf204720q.
Liu, R., J. Cheng, and H. Wu. 2019. Discovery of food-derived dipep-
tidyl peptidase IV inhibitory peptides: A review. International
Journal of Molecular Sciences 20 (3):1–22.
Liu, L., Y. Wang, C. Peng, and J. Wang. 2013. Optimization of the
preparation of sh protein anti-obesity hydrolysates using response
surface methodology. International Journal of Molecular Sciences 14
(2):3124–39. doi: 10.3390/ijms14023124.
Lutz, T., S. Tschudy, P. Rushing, and E. Scharrer. 2000. Amylin recep-
tors mediate the anorectic action of salmon calcitonin (sCT).
Peptides 21 (2):233–8. doi: 10.1016/s0196-9781(99)00208-9.
Macotela, Y., B. Emanuelli, A. M. Bång, D. O. Espinoza, J. Boucher,
K. Beebe, W. Gall, and C. R. Kahn. 2011. Dietary leucine-an envi-
ronmental modier of insulin resistance acting on multiple levels
of metabolism. PLoS One. 6 (6):e21187–13. doi: 10.1371/journal.
pone.0021187.
Madani, Z., K. Louchami, A. Sener, W. J. Malaisse, and D. Ait Yahia.
2012. Dietary sardine protein lowers insulin resistance, leptin and
TNF-α and benecially aects adipose tissue oxidative stress in rats
with fructose-induced metabolic metabolic syndrome. International
Journal of Molecular Medicine 29 (2):311–8. doi: 10.3892/
ijmm.2011.836.
Madani, Z., A. Sener, W. J. Malaisse, and A. Y. Dalila. 2015. Sardine
protein diet increases plasma glucagon-like peptide-1 levels and
prevents tissue oxidative stress in rats fed a high-fructose diet.
Molecular Medicine Reports 12 (5):7017–26. doi: 10.3892/
mmr.2015.4324.
Maeda, H., R. Hosomi, M. Koizumi, Y. Toda, M. Mitsui, and K.
Fukunaga. 2015. Dietary cod protein decreases triacylglycerol ac-
cumulation and fatty acid desaturase indices in the liver of obese
type-2 diabetic KK-Ay mice. Journal of Functional Foods 14:87–94.
doi: 10.1016/j.j.2015.01.038.
Mansour, A., M. R. Mohajeri-Tehrani, M. Qorbani, R. Heshmat, B.
Larijani, and S. Hosseini. 2015. Eect of glutamine supplementation
on cardiovascular risk factors in patients with type 2 diabetes.
Nutrition 31 (1):119–26. doi: 10.1016/j.nut.2014.05.014.
Martinez-Alvarez, O., L. Guimas, C. Delannoy, and M. Fouchereau-Peron.
2007. Occurrence of a CGRP-like molecule in siki (Centroscymnus
coelolepsis) hydrolysate of industrial origin. Journal of Agricultural
and Food Chemistry 55 (14):5469–75. doi: 10.1021/jf070302l.
Martínez-Alvarez, O., R. Ravallec, B. Cudennec, L. Guimas, C.
Delannoy, and M. Fouchereau-Peron. 2012. Eect of daily gavage
with a collagen hydrolysate containing calcitonin gene-related pep-
tide (CGRP)-like molecules on plasma CGRP-levels in rats. Journal
of Functional Foods 4 (4):767–75. doi: 10.1016/j.j.2012.05.004.
Matsui, T., T. Oki, and Y. Osajima. 1999. Isolation and identication
of peptidic α-glucosidase inhibitors derived from sardine muscle
hydrolyzate. Zeitschri Für Naturforschung C 54 (3–4):259–63. doi:
10.1515/znc-1999-3-417.
Matsuyama-Yokono, A., A. Tahara, R. Nakano, Y. Someya, K. Shiraki,
M. Hayakawa, and M. Shibasaki. 2009. Antidiabetic eects of di-
peptidyl peptidase-IV inhibitors and sulfonylureas in
streptozotocin-nicotinamide-induced mildly diabetic mice.
Metabolism: clinical and Experimental 58 (3):379–86. doi: 10.1016/j.
metabol.2008.10.012.
McKnight, J. R., M. C. Sattereld, W. S. Jobgen, S. B. Smith, T. E.
Spencer, C. J. Meininger, C. J. McNeal, and G. Wu. 2010. Benecial
eects of L-arginine on reducing obesity: Potential mechanisms and
important implications for human health. Amino Acids 39 (2):349–
57. doi: 10.1007/s00726-010-0598-z.
Medenieks, L, and T. Vasiljevic. 2008. Underutilised sh as sources of
bioactive peptides with potential health benets. Food Australia 60
(12):581–8.
Messerli, F. H., B. D. Nunez, H. O. Ventura, and D. W. Snyder. 1987.
Overweight and sudden death. Increased ventricular ectopy in car-
diopathy of obesity. Archives of Internal Medicine 147 (10):1725–8.
doi: 10.1001/archinte.147.10.1725.
Michishita, T., S. Kobayashi, T. Katsuya, T. Ogihara, and K. Kawabuchi.
2010. Evaluation of the antiobesity eects of an amino acid mixture
and conjugated linoleic acid on exercising healthy overweight hu-
mans: A randomized, double-blind, placebo-controlled trial. Journal
46 S. ABACHI ETAL.
of International Medical Research 38 (3):844–59. doi:
10.1177/147323001003800311.
Miyake, T., M. Abe, S. Furukawa, Y. Tokumoto, K. Toshimitsu, T.
Ueda, S. Yamamoto, M. Hirooka, T. Kumagi, Y. Hiasa, et al. 2012.
Long-term branched-chain amino acid supplementation improves
glucose tolerance in patients with nonalcoholic steatohepatitis-related
cirrhosis. Internal Medicine 51 (16):2151–5. doi: 10.2169/internal-
medicine.51.7578.
Mizushige, T., M. Komiya, M. Onda, K. Uchida, K. Hayamizu, and Y.
Kabuyama. 2017. Fish protein hydrolysate exhibits anti-obesity ac-
tivity and reduces hypothalamic neuropeptide Y and agouti-related
protein mRNA expressions in rats. Biomedical Research 38 (6):351–
7. doi: 10.2220/biomedres.38.351.
Moore, J. X., N. Chaudhary, and T. Akinyemiju. 2017. Peer reviewed:
Metabolic syndrome prevalence by race/ethnicity and sex in the
United States, National Health and Nutrition Examination Survey,
1988–2012. Preventing Chronic Disease 14:E24. https://www.cdc.gov/
pcd.
Mora, L, and M. Hayes. 2015. Cardioprotective cryptides derived from
sh and other food sources: Generation, application, and future
markets. Journal of Agricultural and Food Chemistry 63 (5):1319–31.
doi: 10.1021/jf505019z.
Morita, T., A. Oh-Hashi, K. Takei, M. Ikai, S. Kasaoka, and S. Kiriyama.
1997. Cholesterol-lowering eects of soybean, potato and rice pro-
teins depend on their low methionine contents in rats fed a
cholesterol-free puried diet. e Journal of Nutrition 127 (3):470–
7. doi: 10.1093/jn/127.3.470.
Mortensen, L. S., M. L. Hartvigsen, L. J. Brader, A. Astrup, J.
Schrezenmeir, J. J. Holst, C. omsen, and K. Hermansen. 2009.
Dierential eects of protein quality on postprandial lipemia in
response to a fat-rich meal in type 2 diabetes: Comparison of whey,
casein, gluten, and cod protein. e American Journal of Clinical
Nutrition 90 (1):41–8. doi: 10.3945/ajcn.2008.27281.
Myrmel, L. S., K. R. Fauske, E. Fjaere, A. Bernhard, U. Liisberg, A.
E. Hasselberg, J. Øyen, K. Kristiansen, and L. Madsen. 2019. e
impact of dierent animal-derived protein sources on adiposity and
glucose homeostasis during ad libitum feeding and energy restric-
tion in already obese mice. Nutrients 11 (5):1153–19. doi: 10.3390/
nu11051153.
Nagao, K, and T. Yanagita. 2008. Bioactive lipids in metabolic syn-
drome. Progress in Lipid Research 47 (2):127–46. doi: 10.1016/j.
plipres.2007.12.002.
Naini, A. E., M. Sadeghi, M. Mortazavi, M. Moghadasi, and A. A.
Harandi. 2012. Oral carnitine supplementation for dyslipidemia in
chronic hemodialysis patients. Saudi Journal of Kidney Diseases and
Transplantation: An Ocial Publication of the Saudi Center for Organ
Transplantation, Saudi Arabia 23 (3):484–8.
Nasri, R., O. Abdelhedi, I. Jemil, I. B. Amor, A. Elfeki, J. Gargouri,
A. Boualga, M. Karra-Châabouni, and M. Nasri. 2018. Preventive
eect of goby sh protein hydrolysates on hyperlipidemia and car-
diovascular disease in Wistar rats fed a high-fat/fructose diet. RSC
Advances 8 (17):9383–93. doi: 10.1039/C7RA13102J.
Nasri, R., O. Abdelhedi, I. Jemil, I. Daoued, K. Hamden, C. Kallel, A.
Elfeki, M. Lamri-Senhadji, A. Boualga, M. Nasri, et al. 2015.
Ameliorating effects of goby fish protein hydrolysates on
high-fat-high-fructose diet-induced hyperglycemia, oxidative stress
and deterioration of kidney function in rats. Chemico-Biological
Interactions 242:71–80. doi: 10.1016/j.cbi.2015.08.003.
Natsir, H., S. Dali, and A. Arif. 2019. Activity and kinetics of
α-glucosidase inhibition by collagen hydrolysate from unnus al-
bacares bone. Journal of Physics: Conference Series. 1341:032015.
Nesse, K. O., A. Nagalakshmi, P. Marimuthu, M. Singh, P. J. Bhetariya,
M. Ho, and R. R. Simon. 2014. Safety evaluation of sh protein
hydrolysate supplementation in malnourished children. Regulatory
Toxicology and Pharmacology: RTP 69 (1):1–6. doi: 10.1016/j.
yrtph.2014.02.009.
Neves, A. C., P. A. Harnedy, M. B. OKeee, and R. J. FitzGerald.
2017. Bioactive peptides from Atlantic salmon (Salmo salar) with
angiotensin converting enzyme and dipeptidyl peptidase IV inhib-
itory, and antioxidant activities. Food Chemistry 218:396–405. doi:
10.1016/j.foodchem.2016.09.053.
Newgard, C. B., J. An, J. R. Bain, M. J. Muehlbauer, R. D. Stevens, L.
F. Lien, A. M. Haqq, S. H. Shah, M. Arlotto, C. A. Slentz, et al.
2009. A branched-chain amino acid-related metabolic signature that
dierentiates obese and lean humans and contributes to insulin
resistance. Cell Metabolism 9 (4):311–26. doi: 10.1016/j.
cmet.2009.02.002.
Nishida, Y., Y. Ide, M. Okada, T. Otsuka, Y. Eguchi, I. Ozaki, K.
Tanaka, and T. Mizuta. 2017. Eects of home‐based exercise and
branched‐chain amino acid supplementation on aerobic capacity
and glycemic control in patients with cirrhosis. Hepatology Research
47 (3):E193–E200. doi: 10.1111/hepr.12748.
Nishimura, J., T. Masaki, M. Arakawa, M. Seike, and H. Yoshimatsu.
2010. Isoleucine prevents the accumulation of tissue triglycerides
and upregulates the expression of PPARα and uncoupling protein
in diet-induced obese mice. e Journal of Nutrition 140 (3):496–
500. doi: 10.3945/jn.109.108977.
Nobile, V., E. Duclos, A. Michelotti, G. Bizzaro, M. Negro, and F.
Soisson. 2016. Supplementation with a sh protein hydrolysate
(Micromesistius poutassou): Eects on body weight, body compo-
sition, and CCK/GLP-1 secretion. Food & Nutrition Research 60
(1):29857–9. doi: 10.3402/fnr.v60.29857.
Nongonierma, A. B, and R. J. FitzGerald. 2013. Inhibition of dipepti-
dyl peptidase IV (DPP-IV) by proline containing casein-derived
peptides. Journal of Functional Foods 5 (4):1909–17. doi: 10.1016/j.
j.2013.09.012.
Nongonierma, A. B, and R. J. FitzGerald. 2014. An in silico model to
predict the potential of dietary proteins as sources of dipeptidyl
peptidase IV (DPP-IV) inhibitory peptides. Food Chemistry 165:489–
98. doi: 10.1016/j.foodchem.2014.05.090.
Oda, H. 2006. Functions of sulfur-containing amino acids in lipid
metabolism. e Journal of Nutrition 136 (6 Suppl):1666S–9S. doi:
10.1093/jn/136.6.1666S.
Opara, E. C., A. Petro, A. Tevrizian, M. N. Feinglos, and R. S. Surwit.
1996. L-glutamine supplementation of a high fat diet reduces body
weight and attenuates hyperglycemia and hyperinsulinemia in
C57BL/6J mice. e Journal of Nutrition 126 (1):273–9. doi: 10.1093/
jn/126.1.273.
Otani, L., T. Ninomiya, M. Murakami, K. Osajima, H. Kato, and T.
Murakami. 2009. Sardine peptide with angiotensin I-converting
enzyme inhibitory activity improves glucose tolerance in stroke-prone
spontaneously hypertensive rats. Bioscience, Biotechnology, and
Biochemistry 73 (10):2203–9. doi: 10.1271/bbb.90311.
Ouellet, V., J. Marois, S. J. Weisnagel, and H. Jacques. 2007. Dietary
cod protein improves insulin sensitivity in insulin-resistant men
and women: A randomized controlled trial. Diabetes Care 30
(11):2816–21. doi: 10.2337/dc07-0273.
Ouellet, V., S. J. Weisnagel, J. Marois, J. Bergeron, P. Julien, R. Gougeon,
A. Tchernof, B. J. Holub, and H. Jacques. 2008. Dietary cod protein
reduces plasma C-reactive protein in insulin-resistant men and
women. e Journal of Nutrition 138 (12):2386–91. doi: 10.3945/
jn.108.092346.
Pal, S, and A. K. Chatterjee. 2004. Protective eect of methionine
supplementation on arsenic-induced alteration of glucose homeo-
stasis. Food and Chemical Toxicology: An International Journal
Published for the British Industrial Biological Research Association
42 (5):737–42. doi: 10.1016/j.fct.2003.12.009.
Pampanin, D. M., E. Larssen, F. Provan, M. Sivertsvik, P. Ruo, and
M. O. Sydnes. 2012. Detection of small bioactive peptides from
Atlantic herring (Clupea harengus L.). Peptides 34 (2):423–6. doi:
10.1016/j.peptides.2012.02.005.
Paneni, F., S. Costantino, and F. Cosentino. 2014. Insulin resistance,
diabetes, and cardiovascular risk. Current Atherosclerosis Reports 16
(7):1–8. doi: 10.1007/s11883-014-0419-z.
Parikh, R. M, and V. Mohan. 2012. Changing denitions of metabol-
ic syndrome. Indian Journal of Endocrinology and Metabolism 16
(1):7–12. doi: 10.4103/2230-8210.91175.
Parolini, C., R. Vik, M. Busnelli, B. Bjørndal, S. Holm, T. Brattelid, S.
Manzini, G. S. Ganzetti, F. Dellera, B. Halvorsen, et al. 2014. A
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 47
salmon protein hydrolysate exerts lipid-independent
anti-atherosclerotic activity in ApoE-decient mice. PLoS One. 9
(5):e97598–9. doi: 10.1371/journal.pone.0097598.
Parra, D., N. M. Bandarra, M. Kiely, I. orsdottir, and J. A. Martínez.
2007. Impact of sh intake on oxidative stress when included into
a moderate energy-restricted program to treat obesity. European
Journal of Nutrition 46 (8):460–7. doi: 10.1007/s00394-007-0686-3.
Parthsarathy, V., C. M. McLaughlin, P. A. Harnedy, P. J. Allsopp, W.
Crowe, E. M. McSorley, R. J. FitzGerald, and F. P. O’harte. 2019.
Boarsh (Capros aper) protein hydrolysate has potent insulinotropic
and GLP‐1 secretory activity in vitro and acute glucose lowering
eects in mice. International Journal of Food Science & Technology
54 (1):271–81. doi: 10.1111/ijfs.13975.
Pérez-Gálvez, R., P. J. García-Moreno, R. Morales-Medina, A. Guadix,
and E. M. Guadix. 2015. Bile acid binding capacity of sh protein
hydrolysates from discard species of the West Mediterranean Sea.
Food & Function 6 (4):1261–7. doi: 10.1039/c4fo01171f.
Fu, W. J., T. E. Haynes, R. Kohli, J. Hu, W. Shi, T. E. Spencer, R. J.
Carroll, C. J. Meininger, and G. Wu. 2005. Dietary L-arginine sup-
plementation reduces fat mass in Zucker diabetic fatty rats. e
Journal of Nutrition 135 (4):714–21. doi: 10.1093/jn/135.4.714.
Petzke, K., A. Freudenberg, and S. Klaus. 2014. Beyond the role of
dietary protein and amino acids in the prevention of diet-induced
obesity. International Journal of Molecular Sciences 15 (1):1374–91.
doi: 10.3390/ijms15011374.
Picard-Deland, É., C. Lavigne, J. Marois, J. Bisson, S. J. Weisnagel, A.
Marette, B. Holub, E. Chu, J. Frohlich, J. S. Hill, et al. 2012. Dietary
supplementation with sh gelatine modies nutrient intake and
leads to sex-dependent responses in TAG and C-reactive protein
levels of insulin-resistant subjects. Journal of Nutritional Science
1:1–10. doi: 10.1017/jns.2012.13.
Pilon, G., J. Ruzzin, L.-E. Rioux, C. Lavigne, P. J. White, L. Frøyland,
H. Jacques, P. Bryl, L. Beaulieu, and A. Marette. 2011. Dierential
eects of various sh proteins in altering body weight, adiposity,
inammatory status, and insulin sensitivity in high-fat-fed rats.
Metabolism: Clinical and Experimental 60 (8):1122–30. doi: 10.1016/j.
metabol.2010.12.005.
Pospisilik, J. A., J. Martin, T. Doty, J. A. Ehses, N. Pamir, F. C. Lynn,
S. Piteau, H.-U. Demuth, C. H. McIntosh, and R. A. Pederson. 2003.
Dipeptidyl peptidase IV inhibitor treatment stimulates beta-cell
survival and islet neogenesis in streptozotocin-induced diabetic rats
. Diabetes 52 (3):741–50. doi: 10.2337/diabetes.52.3.741.
Power, O., A. B. Nongonierma, P. Jakeman, and R. J. Fitzgerald. 2014.
Food protein hydrolysates as a source of dipeptidyl peptidase IV
inhibitory peptides for the management of type 2 diabetes.
Proceedings of the Nutrition Society 73 (1):34–46. doi: 10.1017/
S0029665113003601.
Qin, L. Q., P. Xun, D. Bujnowski, M. L. Daviglus, L. Van Horn, J.
Stamler, and K. He. 2011. Higher branched-chain amino acid intake
is associated with a lower prevalence of being overweight or obese
in middle-aged East Asian and Western adults. e Journal of
Nutrition 141 (2):249–54. doi: 10.3945/jn.110.128520.
Raal, F. J., and R. D. Santos. 2012. Homozygous familial hypercholes-
terolemia: Current perspectives on diagnosis and treatment.
Atherosclerosis 223 (2):262–8. doi: 10.1016/j.atherosclero-
sis.2012.02.019.
Rajamohan, T, and P. Kurup. 1990. Antiatherogenic eect of a low
lysine: Arginine ratio of protein involves alteration in the aortic
glycosamihoglycans and glycoproteins. Journal of Biosciences 15
(4):305–11. doi: 10.1007/BF02702672.
Raksha, N. G., P. Y. Potalitsyn, A. V. Yurchenko, T. I. Halenova, O.
M. Savchuk, and L. I. Ostapchenko. 2018. Prevention of diet-induced
obesity in rats by oral application of collagen fragments. Archives
of Biological Sciences 70 (1):77–86. doi: 10.2298/ABS170401027R.
Ramel, A., M. T. Jonsdottir, and I. orsdottir. 2009. Consumption of cod
and weight loss in young overweight and obese adults on an energy
reduced diet for 8-weeks. Nutrition, Metabolism and Cardiovascular
Diseases 19 (10):690–6. doi: 10.1016/j.numecd.2008.12.013.
Ramel, A., D. Parra, J. A. Martinéz, M. Kiely, and I. orsdottir. 2009.
Eects of seafood consumption and weight loss on fasting leptin
and ghrelin concentrations in overweight and obese European young
adults. European Journal of Nutrition 48 (2):107–14. doi: 10.1007/
s00394-008-0769-9.
Rehfeld, J. F. 2017. Cholecystokinin—from local gut hormone to ubiq-
uitous messenger. Frontiers in Endocrinology 8:1–8. doi: 10.3389/
fendo.2017.00047.
Reimann, F., L. Williams, G. da Silva Xavier, G. Rutter, and F. Gribble.
2004. Glutamine potently stimulates glucagon-like peptide-1 secre-
tion from GLUTag cells. Diabetologia 47 (9):1592–601. doi: 10.1007/
s00125-004-1498-0.
Ricci-Cabello, I., M. Olalla Herrera, and R. Artacho. 2012. Possible
role of milk-derived bioactive peptides in the treatment and pre-
vention of metabolic syndrome. Nutrition Reviews 70 (4):241–55.
doi: 10.1111/j.1753-4887.2011.00448.x.
Roblet, C., M. J. Akhtar, S. Mikhaylin, G. Pilon, T. Gill, A. Marette,
and L. Bazinet. 2016. Enhancement of glucose uptake in muscular
cell by peptide fractions separated by electrodialysis with ltration
membrane from salmon frame protein hydrolysate. Journal of
Functional Foods 22:337–46. doi: 10.1016/j.j.2016.01.003.
Saito, M., C. Kiyose, T. Higuchi, N. Uchida, and H. Suzuki. 2009.
Eect of collagen hydrolysates from salmon and trout skins on the
lipid prole in rats. Journal of Agricultural and Food Chemistry 57
(21):10477–82. doi: 10.1021/jf902355m.
Samocha-Bonet, D., D. J. Chisholm, F. M. Gribble, A. C. Coster, K.
H. Carpenter, G. R. Jones, J. J. Holst, and J. R. Greeneld. 2014.
Glycemic eects and safety of L-Glutamine supplementation with
or without sitagliptin in type 2 diabetes patients—A randomized
study. PLoS One. 9 (11):e113366–7. doi: 10.1371/journal.
pone.0113366.
Samocha-Bonet, D., O. Wong, E.-L. Synnott, N. Piyaratna, A.
Douglas, F. M. Gribble, J. J. Holst, D. J. Chisholm, and J. R.
Greenfield. 2011. Glutamine reduces postprandial glycemia and
augments the glucagon-like peptide-1 response in type 2 diabetes
patients. The Journal of Nutrition 141 (7):1233–8. doi: 10.3945/
jn.111.139824.
Santoso, U., S. Ishikawa, and K. Tanaka. 2010. Eect of fermented
chub mackerel extract on lipid metabolism of diabetic rats. Journal
of the Indonesian Tropical Animal Agriculture 35 (3):158–64. doi:
10.14710/jitaa.35.3.158-164.
Santos-Silva, J. C., R. A. Ribeiro, J. F. Vettorazzi, E. Irles, S. Rickli, P.
C. Borck, P. M. Porciuncula, I. Quesada, A. Nadal, A. C. Boschero,
etal. 2015. Taurine supplementation ameliorates glucose homeosta-
sis, prevents insulin and glucagon hypersecretion, and controls β,
α, and δ-cell masses in genetic obese mice. Amino Acids 47
(8):1533–48. doi: 10.1007/s00726-015-1988-z.
Sasaoka, Y., H. Kishimura, S. Adachi, and Y. Takagi. 2018. Collagen
peptides derived from the triple helical region of sturgeon collagen
improve glucose tolerance in normal mice. Journal of Food
Biochemistry 42 (2):e12478–20. doi: 10.1111/jc.12478.
Sattereld, M. C., K. A. Dunlap, D. H. Keisler, F. W. Bazer, and G.
Wu. 2012. Arginine nutrition and fetal brown adipose tissue devel-
opment in diet-induced obese sheep. Amino Acids. 43 (4):1593–603.
doi: 10.1007/s00726-012-1235-9.
Scheen, A. J, and L. F. Van Gaal. 2014. Combating the dual burden:
erapeutic targeting of common pathways in obesity and type 2
diabetes. e Lancet. Diabetes & Endocrinology 2 (11):911–22. doi:
10.1016/S2213-8587(14)70004-X.
Schulze, F., S. Glos, D. Petruschka, C. Altenburg, R. Maas, R. Benndorf,
E. Schwedhelm, U. Beil, and R. H. Böger. 2009. L-Arginine enhanc-
es the triglyceride-lowering eect of simvastatin in patients with
elevated plasma triglycerides. Nutrition Research (New York, NY) 29
(5):291–7. doi: 10.1016/j.nutres.2009.04.004.
Selvin, E., M. W. Stees, H. Zhu, K. Matsushita, L. Wagenknecht, J.
Pankow, J. Coresh, and F. L. Brancati. 2010. Glycated hemoglobin,
diabetes, and cardiovascular risk in nondiabetic adults. New England
Journal of Medicine 362 (9):800–11. doi: 10.1056/NEJMoa0908359.
Shemesh, E, and B. Zafrir. 2019. Hypertriglyceridemia-related pancre-
atitis in patients with type 2 diabetes: Links and risks. Diabetes,
Metabolic Syndrome and Obesity: Targets and erapy 12:2041–52.
doi: 10.2147/DMSO.S188856.
48 S. ABACHI ETAL.
Shukla, A., A. Bettzieche, F. Hirche, C. Brandsch, G. I. Stangl, and K.
Eder. 2006. Dietary sh protein alters blood lipid concentrations
and hepatic genes involved in cholesterol homeostasis in the rat
model. British Journal of Nutrition 96 (4):674–82.
Siala, R., A. Khabir, I. Lassoued, O. Abdelhedi, A. Elfeki, T. Vallaeys,
and M. Nasri. 2016. Functional and antioxidant properties of pro-
tein hydrolysates from grey triggersh muscle and in vivo evaluation
of hypoglycemic and hypolipidemic activities. Journal of Applied &
Environmental Microbiology 4 (6):105–19.
Sila, A., O. M. Alvarez, A. Haddar, F. Frikha, P. Dhulster, N.
Nedjar-Arroume, and A. Bougatef. 2016. Purication, identication
and structural modelling of DPP-IV inhibiting peptides from barbel
protein hydrolysate. Journal of Chromatography. B, Analytical
Technologies in the Biomedical and Life Sciences 1008:260–9. doi:
10.1016/j.jchromb.2015.11.054.
Sila, A., O. Martinez-Alvarez, A. Haddar, M. C. Gómez-Guillén, M.
Nasri, M. P. Montero, and A. Bougatef. 2015. Recovery, viscoelastic
and functional properties of Barbel skin gelatine: Investigation of
anti-DPP-IV and anti-prolyl endopeptidase activities of generated
gelatine polypeptides. Food Chemistry 168:478–86. doi: 10.1016/j.
foodchem.2014.07.086.
Slizyte, R., K. Rommi, R. Mozuraityte, P. Eck, K. Five, and T. Rustad.
2016. Bioactivities of sh protein hydrolysates from defatted salm-
on backbones. Biotechnology Reports (Amsterdam, Netherlands)
11:99–109. doi: 10.1016/j.btre.2016.08.003.
Solerte, S. B., C. Gazzaruso, N. Schifino, E. Locatelli, T. Destro, G.
Ceresini, E. Ferrari, and M. Fioravanti. 2004. Metabolic effects
of orally administered amino acid mixture in elderly subjects
with poorly controlled type 2 diabetes mellitus. The American
Journal of Cardiology 93 (8):23–9. doi: 10.1016/j.amj-
card.2003.11.006.
Spielmann, J., H. Kluge, G. Stangl, and K. Eder. 2009. Hypolipidaemic
eects of potato protein and sh protein in pigs. Journal of Animal
Physiology and Animal Nutrition 93 (4):400–9. doi:
10.1111/j.1439-0396.2008.00819.x.
Sulochana, K., R. Punitham, and S. Ramakrishnan. 1998. Benecial
eect of lysine and amino acids on cataractogenesis in experimen-
tal diabetes through possible antiglycation of lens proteins.
Experimental Eye Research 67 (5):597–601. doi: 10.1006/
exer.1998.0547.
Tak, Y. J., Y. J. Kim, J. G. Lee, Y.-H. Yi, Y. H. Cho, G. H. Kang, and
S. Y. Lee. 2019. Eect of oral ingestion of low-molecular collagen
peptides derived from skate (Raja Kenojei) skin on body fat in
overweight adults: A randomized, double-blind, placebo-controlled
trial. Marine Drugs 17 (3):157–12. doi: 10.3390/md17030157.
Takeshita, Y., T. Takamura, Y. Kita, H. Ando, T. Ueda, K. Kato, H.
Misu, H. Sunagozaka, Y. Sakai, T. Yamashita, et al. 2012. Benecial
eect of branched-chain amino acid supplementation on glycemic
control in chronic hepatitis C patients with insulin resistance:
Implications for type 2 diabetes. Metabolism: clinical and Experimental
61 (10):1388–94. doi: 10.1016/j.metabol.2012.03.011.
Tan, B., Y. Yin, Z. Liu, X. Li, H. Xu, X. Kong, R. Huang, W. Tang, I.
Shinzato, S. B. Smith, et al. 2009. Dietary L-arginine supplementa-
tion increases muscle gain and reduces body fat mass in
growing-nishing pigs. Amino Acids. 37 (1):169–75. doi: 10.1007/
s00726-008-0148-0.
Tastesen, H. S., A. K. Rønnevik, K. Borkowski, L. Madsen, K.
Kristiansen, and B. Liaset. 2014. A mixture of cod and scallop
protein reduces adiposity and improves glucose tolerance in high-fat
fed male C57BL/6J mice. PLoS One. 9 (11):e112859–12. doi: 10.1371/
journal.pone.0112859.
Tenner, T. E. X. J. Zhang, and J. B. Lombardini. 2003. Hypoglycemic
eects of taurine in the alloxan-treated rabbit, a model for type 1
diabetes. Advances in Experimental Medicine and Biology 526:97–104.
Thompson, P. D., G. Panza, A. Zaleski, and B. Taylor. 2016.
Statin-associated side eects. Journal of the American College of
Cardiology 67 (20):2395–410. doi: 10.1016/j.jacc.2016.02.071.
orsdottir, I., H. Tomasson, I. Gunnarsdottir, E. Gisladottir, M. Kiely,
M. D. Parra, N. M. Bandarra, G. Schaafsma, and J. A. Martinez.
2007. Randomized trial of weight-loss-diets for young adults vary-
ing in sh and sh oil content. International Journal of Obesity
(2005) 31 (10):1560–6. doi: 10.1038/sj.ijo.0803643.
Tometsuka, C., Y-i. Koyama, T. Ishijima, T. Toyoda, M. Teranishi, K.
Takehana, K. Abe, and Y. Nakai. 2017. Collagen peptide ingestion
alters lipid metabolism-related gene expression and the unfolded
protein response in mouse liver. British Journal of Nutrition 117
(1):1–11. doi: 10.1017/S0007114516004384.
Tørris, C., M. C. Småstuen, and M. Molin. 2018. Nutrients in sh and
possible associations with cardiovascular disease risk factors in met-
abolic syndrome. Nutrients 10 (7):952–17. doi: 10.3390/nu10070952.
Tripathi, P., M. Misra, and S. Pandey. 2012. Role of l-Arginine on
dyslipidemic conditions of acute myocardial infarction patients.
Indian Journal of Clinical Biochemistry: IJCB 27 (3):296–9. doi:
10.1007/s12291-012-0188-9.
Triplitt, C. 2006. Drug interactions of medications commonly used in
diabetes. Diabetes Spectrum 19 (4):202–11. doi: 10.2337/dias-
pect.19.4.202.
Tulipano, G., V. Sibilia, A. M. Caroli, and D. Cocchi. 2011. Whey
proteins as source of dipeptidyl dipeptidase IV (dipeptidyl
peptidase-4) inhibitors. Peptides 32 (4):835–8. doi: 10.1016/j.pep-
tides.2011.01.002.
Ueda, K., Y. Nakamura, M. Yamaguchi, T. Mori, M. Uchida, and S.
Fujita. 2016. Amino acid mixture enriched with arginine, alanine,
and phenylalanine stimulates fat metabolism during exercise.
International Journal of Sport Nutrition and Exercise Metabolism 26
(1):46–54. doi: 10.1123/ijsnem.2015-0137.
Uhe, A. M., G. R. Collier, and K. O’Dea. 1992. A comparison of the
eects of beef, chicken and sh protein on satiety and amino acid
proles in lean male subjects. e Journal of Nutrition 122 (3):467–
72. doi: 10.1093/jn/122.3.467.
Uhlig, T., T. Kyprianou, F. G. Martinelli, C. A. Oppici, D. Heiligers,
D. Hills, X. R. Calvo, and P. Verhaert. 2014. e emergence of
peptides in the pharmaceutical business: From exploration to ex-
ploitation. EuPA Open Proteomics 4:58–69. doi: 10.1016/j.eu-
prot.2014.05.003.
van Loon, L. J., M. Kruijshoop, P. P. Menheere, A. J. Wagenmakers,
W. H. Saris, and H. A. Keizer. 2003. Amino acid ingestion strong-
ly enhances insulin secretion in patients with long-term type 2
diabetes. Diabetes Care 26 (3):625–30. doi: 10.2337/diacare.26.3.625.
Van Nguyen, M., I. Rønnestad, L. Buttle, H. Van Lai, and M. Espe.
2014. Imbalanced lysine to arginine ratios reduced performance in
juvenile cobia (R achycentron canadum) fed high plant protein
diets. Aquaculture Nutrition 20 (1):25–35. doi: 10.1111/anu.12043.
Vega-López, S., N. R. Matthan, L. M. Ausman, S. V. Harding, T. C.
Rideout, M. Ai, S. Otokozawa, A. Freed, J. T. Kuvin, P. J. Jones,
et al. 2010. Altering dietary lysine:arginine ratio has little eect on
cardiovascular risk factors and vascular reactivity in moderately
hypercholesterolemic adults. Atherosclerosis 210 (2):555–62. doi:
10.1016/j.atherosclerosis.2009.12.002.
Verreijen, A. M., Verlaan, S.Engberink, M. F.Swinkels, S. d.Vogel-van
den Bosch, J, and Weijs, P. J. 2015. A high whey protein–, leucine-,
and vitamin D–enriched supplement preserves muscle mass during
intentional weight loss in obese older adults: A double-blind ran-
domized controlled trial. e American Journal of Clinical Nutrition
101 (2):279–86. doi: 10.3945/ajcn.114.090290.
Vidhya, S., R. Ramya, K. Coral, K. Sulochana, and S. Bharathidevi.
2018. Free amino acids hydroxyproline, lysine, and glycine promote
dierentiation of retinal pericytes to adipocytes: A protective role
against proliferative diabetic retinopathy. Experimental Eye Research
173:179–87. doi: 10.1016/j.exer.2018.05.004.
Vik, R., V. Tillander, J. Skorve, T. Vihervaara, K. Ekroos, S. E. Alexson,
R. K. Berge, and B. Bjørndal. 2015. ree dierently generated
salmon protein hydrolysates reveal opposite eects on hepatic lipid
metabolism in mice fed a high-fat diet. Food Chemistry 183:101–10.
doi: 10.1016/j.foodchem.2015.03.011.
Vikøren, L. A., O. K. Nygård, E. Lied, E. Rostrup, and O. A.
Gudbrandsen. 2013. A randomised study on the eects of sh pro-
tein supplement on glucose tolerance, lipids and body composition
in overweight adults. British Journal of Nutrition 109 (4):648–57.
doi: 10.1017/S0007114512001717.
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 49
von Post-Skagegård, M., B. Vessby, and B. Karlström. 2006. Glucose
and insulin responses in healthy women aer intake of composite
meals containing cod-, milk-, and soy protein. European Journal of
Clinical Nutrition 60 (8):949–54. doi: 10.1038/sj.ejcn.1602404.
Wang, T.-Y., C.-H. Hsieh, C.-C. Hung, C.-L. Jao, M.-C. Chen, and K.-C.
Hsu. 2015. Fish skin gelatin hydrolysates as dipeptidyl peptidase IV
inhibitors and glucagon-like peptide-1 stimulators improve glycaemic
control in diabetic rats: A comparison between warm-and cold-water
sh. Journal of Functional Foods 19:330–40. doi: 10.1016/j.j.2015.09.037.
Wang, L-j., Y-h. Yu, L-g. Zhang, Y. Wang, N. Niu, Q. Li, and L-m.
Guo. 2008. Taurine rescues vascular endothelial dysfunction in
streptozocin-induced diabetic rats: Correlated with downregulation
of LOX-1 and ICAM-1 expression on aortas. European Journal of
Pharmacology 597 (1–3):75–80. doi: 10.1016/j.ejphar.2008.08.031.
Wang, J., Z. Zhang, X. Pei, R. Yang, and Y. Li. 2010. Eects of marine
collagen peptides on blood glucose and lipid metabolism in hyper-
insulinemic rats. Wei sheng yan jiu=. Wei Sheng Yan Jiu = Journal
of Hygiene Research 39 (2):143–6.
Wang, L., S. Zhou, R. Li, L. Zheng, J. Zhu, S. Hu, and Y. Sun. 2012.
Serum levels of calcitonin gene-related peptide and substance P are
decreased in patients with diabetes mellitus and coronary artery
disease. Journal of International Medical Research 40 (1):134–40.
doi: 10.1177/147323001204000114.
Wergedahl, H., O. A. Gudbrandsen, T. H. Røst, and R. K. Berge. 2009.
Combination of sh oil and sh protein hydrolysate reduces the
plasma cholesterol level with a concurrent increase in hepatic cho-
lesterol level in high-fat-fed Wistar rats. Nutrition (Burbank, Los
Angeles County, CA) 25 (1):98–104. doi: 10.1016/j.nut.2008.07.005.
Wergedahl, H., B. Liaset, O. A. Gudbrandsen, E. Lied, M. Espe, Z.
Muna, S. Mørk, and R. K. Berge. 2004. Fish protein hydrolysate
reduces plasma total cholesterol, increases the proportion of HDL
cholesterol, and lowers acyl-CoA:cholesterol acyltransferase activity
in liver of Zucker rats. e Journal of Nutrition 134 (6):1320–7. doi:
10.1093/jn/134.6.1320.
Woo, M., Y. Song, K.-H. Kang, and J. Noh. 2018. Anti-obesity eects
of collagen peptide derived from skate (Raja kenojei) skin through
regulation of lipid metabolism. Marine Drugs 16 (9):306–12. doi:
10.3390/md16090306.
Wu, G., F. W. Bazer, R. C. Burghardt, G. A. Johnson, S. W. Kim, D.
A. Knabe, P. Li, X. Li, J. R. McKnight, M. C. Sattereld, etal. 2011.
Proline and hydroxyproline metabolism: Implications for animal
and human nutrition. Amino Acids 40 (4):1053–63. doi: 10.1007/
s00726-010-0715-z.
Wu, X., H. Motoshima, K. Mahadev, T. J. Stalker, R. Scalia, and B. J.
Goldstein. 2003. Involvement of AMP-activated protein kinase in
glucose uptake stimulated by the globular domain of adiponectin
in primary rat adipocytes. Diabetes 52 (6):1355–63. doi: 10.2337/
diabetes.52.6.1355.
Xia, E.-Q., S.-S. Zhu, M.-J. He, F. Luo, C.-Z. Fu, and T.-B. Zou. 2017.
Marine peptides as potential agents for the management of type 2
diabetes mellitus—a prospect. Marine Drugs 15 (4):88–16. doi:
10.3390/md15040088.
Xu, H., Y. Mu, Y. Zhang, J. Li, M. Liang, K. Zheng, and Y. Wei. 2016.
Graded levels of sh protein hydrolysate in high plant diets for
turbot (Scophthalmus maximus): eects on growth performance and
lipid accumulation. Aquaculture 454:140–7. doi: 10.1016/j.aquacul-
ture.2015.12.006.
Yahia, D. A., S. Madani, J. Prost, M. Bouchenak, and J. Belleville. 2005.
Fish protein improves blood pressure but alters HDL 2 and HDL
3 composition and tissue lipoprotein lipase activities in sponta-
neously hypertensive rats. European Journal of Nutrition 44 (1):10–
7. doi: 10.1007/s00394-004-0486-y.
Yahia, D. A., S. Madani, E. Prost, J. Prost, M. Bouchenak, and J.
Belleville. 2003. Tissue antioxidant status diers in spontaneously
hypertensive rats fed sh protein or casein. e Journal of Nutrition
133 (2):479–82. doi: 10.1093/jn/133.2.479.
Yang, J., M. Dolinger, G. Ritaccio, J. Mazurkiewicz, D. Conti, X. Zhu,
and Y. Huang. 2012. Leucine stimulates insulin secretion via
down-regulation of surface expression of adrenergic α2A Receptor
through the mTOR (Mammalian Target of Rapamycin) pathway
implication in new-onset diabetes in renal transplantation. Journal
of Biological Chemistry 287 (29):24795–806. doi: 10.1074/jbc.
M112.344259.
Yanni, A. E., D. N. Perrea, and H. A. Yatzidis. 2005. Eect of antia-
therogenic L-aspartate and L-glutamate on serum lipoproteins cho-
lesterol and apolipoproteins A-1 and B in rabbits fed with high
cholesterol diet. Nutrition, Metabolism and Cardiovascular Diseases
15 (3):161–5. doi: 10.1016/j.numecd.2004.06.001.
Yao, K., Y. Duan, F. Li, B. Tan, Y. Hou, G. Wu, and Y. Yin. 2016.
Leucine in obesity: erapeutic prospects. Trends in Pharmacological
Sciences 37 (8):714–27. doi: 10.1016/j.tips.2016.05.004.
Yuan, G., K. Z. Al-Shali, and R. A. Hegele. 2007. Hypertriglyceridemia:
Its etiology, effects and treatment. CMAJ: Canadian Medical
Association Journal = Journal de L’Association Medicale Canadienne
176 (8):1113–20. doi: 10.1503/cmaj.060963.
Zaïr, Y., E. Duclos, B. Housez, C. Vergara, M. Cazaubiel, and F. Soisson.
2014. Evaluation of the satiating properties of a sh protein hydro-
lysate among overweight women: A pilot study. Nutrition & Food
Science 44 (5):389–99. doi: 10.1108/NFS-06-2013-0075.
Zaribaf, F., E. Falahi, F. Barak, M. Heidari, A. Keshteli, A. Yazdannik,
and A. Esmaillzadeh. 2014. Fish consumption is inversely associat-
ed with the metabolic syndrome. European Journal of Clinical
Nutrition 68 (4):474–80. doi: 10.1038/ejcn.2014.5.
Zhang, Y., R. Chen, X. Chen, Z. Zeng, H. Ma, and S. Chen. 2016.
Dipeptidyl peptidase IV-inhibitory peptides derived from Silver Carp
(Hypophthalmichthys molitrix Val.) proteins. Journal of Agricultural
and Food Chemistry 64 (4):831–9. doi: 10.1021/acs.jafc.5b05429.
Zhang, R., J. Chen, X. Jiang, L. Yin, and X. Zhang. 2016. Antioxidant
and hypoglycaemic eects of tilapia skin collagen peptide in mice.
International Journal of Food Science & Technology 51 (10):2157–63.
doi: 10.1111/ijfs.13193.
Zhang, Y., K. Guo, R. E. LeBlanc, D. Loh, G. J. Schwartz, and Y.-H. Yu.
2007. Increasing dietary leucine intake reduces diet-induced obesity
and improves glucose and cholesterol metabolism in mice via multi-
mechanisms. Diabetes 56 (6):1647–54. doi: 10.2337/db07-0123.
Zhang, Y., N. Wang, W. Wang, J. Wang, Z. Zhu, and X. Li. 2016. Molecular
mechanisms of novel peptides from silkworm pupae that inhibit
α-glucosidase. Peptides 76:45–50. doi: 10.1016/j.peptides.2015.12.004.
Zhang, C., Y. Zhang, Z. Wang, S. Chen, and Y. Luo. 2017. Production
and identication of antioxidant and angiotensin-converting enzyme
inhibition and dipeptidyl peptidase IV inhibitory peptides from
bighead carp (Hypophthalmichthys nobilis) muscle hydrolysate.
Journal of Functional Foods 35:224–35. doi: 10.1016/j.j.2017.05.032.
Zheng, K., T. Xu, C. Qian, M. Liang, and X. Wang. 2014. Eect of
low molecular weight sh protein hydrolysate on growth perfor-
mance and IGF‐I expression in J apanese ounder (P aralichthys
olivaceus) fed high plant protein diets. Aquaculture Nutrition 20
(4):372–80. doi: 10.1111/anu.12090.
Zhu, C.-F., G.-Z. Li, H.-B. Peng, Y. Li, F. Zhang, and Y. Chen. 2010.
erapeutic eects of marine collagen peptides on Chinese patients
with type 2 diabetes mellitus and primary hypertension. The
American Journal of the Medical Sciences 340 (5):360–6. doi: 10.1097/
MAJ.0b013e3181edfcf2.
Zhu, C.-F., G.-Z. Li, H.-B. Peng, F. Zhang, Y. Chen, and Y. Li. 2010.
Treatment with marine collagen peptides modulates glucose and
lipid metabolism in Chinese patients with type 2 diabetes mellitus.
Applied Physiology. Nutrition, and Metabolism 35 (6):797–804.
Zhu, C., W. Zhang, J. Liu, B. Mu, F. Zhang, N. Lai, J. Zhou, A. Xu,
and Y. Li. 2017. Marine collagen peptides reduce endothelial cell
injury in diabetic rats by inhibiting apoptosis and the expression
of coupling factor 6 and microparticles. Molecular Medicine Reports
16 (4):3947–57. doi: 10.3892/mmr.2017.7061.
Zhu, C., W. Zhang, B. Mu, F. Zhang, N. Lai, J. Zhou, A. Xu, J. Liu, and
Y. Li. 2017. Eects of marine collagen peptides on glucose metabolism
and insulin resistance in type 2 diabetic rats. Journal of Food Science
and Technology 54 (8):2260–9. doi: 10.1007/s13197-017-2663-z.
Zhu, C.-F., F. Zhu, and J.-X. Zhou. 2013. e intervention eect of
marine collagen peptide on atherosclerosis, cardiac structure and
cardiac function in hypertension patients. Chinese Journal of
Gerontology 33:1741–4.
... Therefore, fish products are potential therapy candidates for prevention and treatment risk factors. 4 However, herein beneficial effects of various fish species on metabolic dysfunction are required for further comprehensively discussed. Mackerel, milkfish, and perch, rich in biologically active amino acids and protein, are three popular fish species in Taiwan. ...
... As most of the fish proteins that are effective anti-Mets are bio-peptides with molecular weights below 1000. 4,17 To investigate why perch has the highest activity, this study further analyzed the molecular weight distribution of perch essences using gel filtration chromatography. The perch essences with molecular weight less than 1.2 kDa accounted for the total protein was 22.6% from the results of protein profile analysis in Table 4. ...
... Many research indicated that bioactive fish proteins or peptides tend to be present as oligo-peptides with low molecular weight and contain 2 to 20 amino acid residues (near 3 kDa). 3,4 This result suggested that although the majority of molecules of perch essences are proteins determined by the colorimetric method as in Table 1, the high inhibitory activity of perch essences on digestive enzymes might be related to the presence of these smaller molecular proteins or peptides with a molecular weight of less than 2.3 kDa. The metabolic health effects of a fish protein depend on several factors, such as the amino acid composition of the protein, the steric structure of the protein, and chemical changes of the protein during processing. ...
Article
Full-text available
The objective of this study was to analyze the protein profile and amino acid composition of perch essence processed by high-pressure extraction compared with mackerel, and milkfish. The inhibitory activities of three essences were determined against digestive enzymes relevant to the metabolic syndrome such as the angiotensin-converting enzyme (ACE), pancreatic lipase, α-amylase, and α-glucosidase. The results showed that perch essence exhibits the highest recovery by using the same extraction method for all three fish species. The amounts of all hydrolyzed amino acids, except histidine, were approximately double in perch compared with the other two fish. The perch essence contained the most abundant soluble protein and amino acids composition related to metabolic syndrome. In vitro anti-hypertensive, hypoglycemic, and anti-obesity potency of perch essence exhibited the highest among the three fishes. Although the amounts of free amino acids were similar in the three fish species, the molecular weight distribution from gel filtration chromatographic analysis indicated that perch essence contained 32.4% of peptides with a molecular weight of less than 2.3 kDa. This study suggested that naturally occurring bioactive peptides in perch essence might potentially serve as a good source of functional food supplements for metabolic syndrome.
... Currently, although some drugs are available to treat or alleviate MetS, most of them are constantly accompanied by undesirable side effects [8]. Noticeably, MetS is believed to be mostly caused by a high-fat or high-sugar diet and deficient physical activity. ...
Article
Full-text available
Pulses, as an important part of the human diet, can act as a source of high-quality plant proteins. Pulse proteins and their hydrolysates have shown promising results in alleviating metabolic syndrome and modulating the gut microbiome. Their bioactivities have become a focus of research, with many new findings added in recent studies. This paper comprehensively reviews the anti-hypertension, anti-hyperglycemia, anti-dyslipidemia and anti-obesity bioactivities of pulse proteins and their hydrolysates in recent in vitro and in vivo studies, which show great potential for the prevention and treatment of metabolic syndrome. In addition, pulse proteins and their hydrolysates can regulate the gut microbiome, which in turn can have a positive impact on the treatment of metabolic syndrome. Furthermore, the beneficial effects of some pulse proteins and their hydrolysates on metabolic syndrome have been supported by clinical studies. This review might provide a reference for the application of pulse proteins and their hydrolysates in functional foods or nutritional supplements for people with metabolic syndrome.
... The peptide molecular weight is of special interest for FPH sensory characteristics, such as bitterness [31][32][33][34][35][36], as well as functional properties, like emulsion, solubility capacity [23,37,38], and bioactivities [25,[39][40][41]. The formation of bitter taste is mainly ascribed to small peptides of less than 1000 Da or up to 8-10 amino acids [31,42]. ...
Article
Full-text available
The rest raw materials of whitefish have great potential for increased utilisation and value creation. Whitefish heads have a high protein content and should be considered a healthy protein source for the growing population’s demands for sustainable protein. In this study, the heads of four different species of whitefish were processed via enzymatic hydrolysis, namely cod (Gadus morhua), cusk (Brosme bromse), haddock (Melanogrammus aeglefinus), and saithe (Pollachius virens), using three commercially available enzymes. Trials were conducted after 0, 3, and 6 months of the frozen storage of heads. A proximate analysis, molecular weight distribution, and protein solubility were evaluated for each of the products. The results show that, although the enzymatic hydrolysis of rest raw materials from different species of whitefish yielded products of slightly different characteristics, this process is viable for the production of high-quality protein from cod, cusk, haddock, and saithe heads. Six months of frozen storage of heads had a minimal effect on the yield and proximate composition of hydrolysates.
... Peptide research is suspected to have a physiological mechanism of action and does not cause side effects, and is strongly suspected to have the potential to prevent the development of pathological processes during COVID-19 by inhibiting the SARS-CoV-2 viral protein, so that it has immunoprotective and bronchoprotective effects on lung cells, and normalizes the state of the immune system and hemostatic system, such as antimicrobial peptides [2]. Moreover, in 2022, researchers have been discovered beneficial effect of fish peptides for drugs degenerative diseases (anti-obesogenic, hypoglycemic, hypercholesterolemic and hypolipidemic) [1,26]. Conventional antibiotic compounds are basically synthetic types of peptides, and resistance to their use are found, so it is necessary to find a new sources of natural peptides (i.e marine peptides), so that they can overcome conventional antibiotic resistance cases. ...
Article
Full-text available
In the pasca of the COVID-19 pandemic, many researchers have focused on finding drugs for the COVID-19. Peptide research is predicted to have a physiological mechanism of action and it does not cause side effects, therefore it is necessary to search for peptide sources. The aim of this study was to characterize peptides extracted from tamban fish ( Sardinella fimbriata ) and fermented products ( Cincalok of Tanjungpinang ). Peptide extract was obtained by maceration extraction using 95% PA (1:5) ethanol solvent, and then it filtered using MWCO 3 kDa and 10 kDa. The amino acid characteristics of the peptides was obtained using HPLC. Peptides of tamban fish had the pH of 7.6 and they are rich of amino acid histidine (890.18 mg/L), in addition to the amino acids glutamate, tyrosine, glycine, leucine, alanine, and aspartic acid. But the peptides of the fermented cincalok product had the pH of 6.4 and they are rich of amino acid leucine (1066.83 mg/L), in addition to the amino acids glutamate, valine, isoleucine, and alanine. Each of these peptides had the molecular weight of 10 kDa. The results of this study can be used as preliminary information to the pharmaceutical development of a new source of peptides.
... Fatty acids play an important role as a source of metabolic energy in fish growth [66]. However, the abnormal lipid metabolism in fish is a risk factor for diabetes, hyperlipidemia, and metabolic diseases [67,68]. In recent years, several studies have demonstrated the positive role of CNE in the treatment of lipid metabolism disorders. ...
Article
Full-text available
Fat greenling (Hexagrammos otakii) is a kind of economic fish that is widely consumed by human, and its intensive farming technology is making important progress. However, high-density farming may cause the occurrence of diseases in H. otakii. Cinnamaldehyde (CNE) is a new feed additive for aquatic animals and has a positive effect on disease resistance. In the study, dietary CNE was evaluated on the growth performance, digestion, immune response, and lipid metabolism of juvenile H. otakii (6.21±0.19 g). Six experimental diets were formulated containing CNE at levels of 0, 200, 400, 600, 800, and 1000 mg/kg for 8 weeks. The percent weight gain (PWG), specific growth rate (SGR), survival (SR), and feeding rate (FR) were significantly increased by including CNE in fish diets regardless of the inclusion level (P
Chapter
Full-text available
This chapter provides an overview of the forthcoming chapters in the book, which aim to comprehensively explore the diverse applications of nanochitosan in revolutionizing and augmenting fisheries and aquaculture practices. The book seeks to elucidate the transformative potential of nanochitosan, emphasizing its roles in enhancing sustainability, productivity, and the overall health of aquatic ecosystems. It serves to establish a foundational understanding by detailing the fundamental properties, synthesis methods, characterization techniques, and safety considerations of nanochitosan relevant to its application in fisheries and aquaculture. The book explores nanochitosan’s pivotal role in addressing diseases among fish and other aquatic species, highlighting its antimicrobial properties and potential as an immune system enhancer in aquaculture. It explores nanochitosan’s effectiveness in water treatment, emphasizing its ability to purify aquatic environments by eliminating contaminants, heavy metals, and pollutants while assessing its environmental sustainability. It thoroughly investigates nanochitosan’s applications in augmenting aquaculture productivity, including optimizing feed formulations, fostering growth, enhancing nutrient absorption, and bolstering overall efficiency. Additionally, it examines novel applications of nanochitosan in areas such as fish disease management, water quality improvement, feed enhancement, fish breeding, and detoxification practices. The book aims to outline future research directions, addressing challenges and facilitating further advancements in the field while advocating for environmentally friendly aquaculture practices supported by innovative nanochitosan applications. The aspect of potential environmental concerns associated with the broad application of nanochitosan was explored to consider regulatory compliance. By encompassing these aspects, the book is poised to be a comprehensive resource for researchers, practitioners, policymakers, and industry stakeholders engaged in fisheries and aquaculture. It aims to deepen the understanding of nanochitosan’s transformative capabilities and its role in shaping sustainable, efficient, and resilient practices within these sectors.
Chapter
The underutilized fishery resources, such as non-commercial species and by-products generated on board and in land (e.g. heads, trimmings, skin and others), are an opportunity to obtain food products for human consumption, in line with the Sustainable Development Goals. Fish contains high-quality protein, with a content on a wet basis normally ranging from 15 to 25%, an adequate amino acid profile and good digestibility. Enzymatic hydrolysis of proteins releases peptides that may exert several physiological effects by interacting with human metabolism. Fish waste materials have been reported as a compelling source of antidiabetic peptides, which could be employed as ingredients to fortify foodstuffs intended for nutraceutical applications. This chapter provides a review of the production and characterization of antidiabetic peptides from fish waste protein, the mechanisms explaining their antidiabetic effect and their potential incorporation in food matrices as functional ingredients.
Article
Full-text available
The severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), which causes the coronavirus disease 2019 (COVID-19), is spreading worldwide. Although the COVID-19 epidemic has passed its peak of transmission, the harm it has caused deserves our attention. Scientists are striving to develop medications that can effectively treat COVID-19 symptoms without causing any adverse reactions. SARS-CoV-2 inhibitory peptides derived from animal proteins have a wide range of functional activities in addition to safety. Identifying animal protein sources is crucial to obtaining SARS-CoV-2 inhibitory peptides from animal sources. This review aims to reveal the mechanisms of action of these peptides on SARS-CoV-2 and the possibility of animal proteins as a material source of SARS-CoV-2 inhibitory peptides. Also, it introduces the utilization of computer-aided design methods, phage display, and drug delivery strategies in the research on SARS-CoV-2 inhibitor peptides from animal proteins. In order to identify new antiviral peptides and boost their efficiency, we recommend investigating the interaction between SARS-CoV-2 inhibitory peptides from animal protein sources and non-structural proteins (Nsps) using a variety of technologies, including computer-aided drug approaches, phage display techniques, and drug delivery techniques. This article provides useful information for the development of novel anti-COVID-19 drugs.
Article
For full use of Skipjack tuna (Katsuwonus pelamis) canning by-products, gelatins (STG) were extracted from tuna skins using acid (STG-A), enzyme (STG-E) and hot water (STG-H) methods with yields of 65.30 ± 1.56%, 39.83 ± 1.61%, and 50.97 ± 1.44%, respectively. Due to the high imino acid contents (228, 216, and 213 residues/1000 residues for STG-A, STG-E, and STG-H, respectively), STG-A showed the highest transparency and gel strength properties. Amino acid analysis, sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), ultraviolet (UV) and Fourier transform infrared spectroscopy (FTIR) analysis confirmed that STG-A, STG-E, and STG-H kept the main structure of type I collagen, but enzyme and hot water extraction methods showed much stronger hydrolysis ability on α and β chains. Moreover, neutrase hydrolysate (STG-AH) of STG-A showed the highest 1,1-diphenyl-2-picrylhydrazyl radical (DPPH•) scavenging activity and the fraction STG-AH-І (<3.5 kDa) from STG-AH prepared by ultrafiltration could significantly protect human skin fibroblasts (HSFBs) against ultraviolet-A (UVA) injury. Furthermore, nineteen peptides with high antioxidant activity were purified from STG-AH-І and identified. These results suggested that STG-AH and its derived antioxidant peptides could serve as potential ingredients applied in health benefiting products for preventing UVA injury.
Article
Full-text available
Biomolecules offering health effects on inflammation, as cause or effect of metabolic syndrome, are the subject of many research studies. Among different classes of marine and terrestrial bioactive compounds, fish-extracted biopeptides are of great importance since they have been proven to modulate inflammation in different experimental settings. The aim of this study was to fractionate and identify Atlantic mackerel (Scomber scombrus) peptide fractions with in vitro immunomodulatory activity in lipopolysaccharide (LPS)-stimulated macrophages. Peptide fractions of mackerel were generated using solid phase extraction (SPE, a chromatographic technique), ultrafiltration (UF, a pressure driven separation technique), and electrodialysis with UF membrane (EDUF, an electrically driven separation technique) under various experimental pH conditions. Results suggested that the separation technique plays an important role in the isolation efficiency of immunomodulatory biopeptides. Hydrophobic (SPE isolated) and charged peptides (EDUF isolated), respectively, had anti-inflammation and pro-inflammation effects, while small size peptides (UF isolated) had no bioactivity. Hydrophobic fraction prevented the LPS-stimulated inflammation, non-cytotoxically, by about 17% at a concentration of 10 µg mL⁻¹ in comparison with the positive control. Furthermore, anionic peptides of EDUF and pH3 as well as unprocessed mackerel hydrolysate induced the production of nitric oxide by about 27% demonstrating pro-inflammatory effects in comparison with the negative control. In conclusion, it is presumed that hydrophobicity and charge of a peptide could be rather important factors for immunomodulation activity of an Atlantic mackerel fraction.
Article
Full-text available
Many types of marine-derived products specifically biopeptides, among other health benefiting properties, such as anti-oxidation, anti-infection, anti-hypertension, can also positively affect the immune system through modulations of its cellular responses hence preventing and/or treating many unhealthy conditions inclusive of the metabolic syndrome and its core risk factors. These bioactivities are not solely defined for specific species and many types of fish, farmed or wild-caught, for example, generally salmon, tilapia, cod, sardine, croaker, and others, exhibit comparable health effects. Immuno-potentiating effects of fish-derived peptides on acute and chronic inflammation as well as its related diseases have repeatedly been documented over recent decades. A vast number of pro- and anti-inflammatory cytokines and chemokines have been the subject of these analyses. However, only handful such as interleukins, prostaglandins, etc. and associated signalling pathways, which could simultaneously play pro- and/or anti-inflammatory roles in the innate and/or adaptive immunity of the host, are commonly tested. In addition to the inflammatory ailments, such as inflammatory bowel syndrome, glomerulonephritis, hepatitis and/or arthritis, many of these cytokines and chemokines are also the cause or the effect of metabolic syndrome and its linked risk factors. To date, numerous reviews on the topics of natural, food, animal, dairy and marine immunoregulatory peptides have been published, yet none explicitly has reviewed fish biopeptides. Therefore, cooperatively this document is prepared with the primary focus on the extraction, isolation and immunomodulating bio-effects of fish extracted peptides on cytokines, which are the crucial elements in development and establishment of metabolic diseases and related risk factors, for example, hypertension, obesity, insulin resistance and others. To extend the knowledge of readers on the topic and its trends, authors will briefly discuss the drug-likeness and the pharmacokinetics, as well as the predicted target proteins of fish immunomodulating peptides.
Article
Full-text available
IntroductionDiabetes is a growing epidemic that imposes a substantial economic burden on healthcare systems. This study aimed to evaluate the cost of managing type 2 diabetes (T2D) with dipeptidyl peptidase 4 inhibitors (DPP4Is) using real-world data.Method This longitudinal study used data from the French EGB (Echantillon Généraliste des Bénéficiaires) database. The annual average direct healthcare cost of treating patients with T2D was calculated 3 years prior and 3 years after initiation of DPP4I therapy. Actual total ambulatory and hospital care expenditure for the 3 years after DPP4I initiation was compared to projected costs. The distribution of costs across all care modalities was assessed over the 6-year period.ResultsAmbulatory and hospital care expenditure data for 919 patients with T2D starting DPP4I therapy alone or in combination in 2013 were analyzed. A total of 526 patients (57.2%) were still being treated with DPP4I 3 years after DPP4I initiation. Regardless of the treatment regimen, the ambulatory and hospital care costs increased above projected costs in the first year following DPP4I initiation, and then declined during the second and third years to levels in line with or below projected values for patients using DPP4Is as an add-on therapy. The increase in total expenditure in the first year following DPP4I initiation and the subsequent decline in costs in the second and third years were both associated with general trends in consumption across all aspects of patient care.Conclusion Despite an initial increase in healthcare expenditure, concomitant with reevaluation of patient care, this study showed that initiation of DPP4Is as an add-on therapy in French patients with T2D was associated with care expenditure that was in line or below predicted values within the 3 years following treatment initiation. Additional studies are required to evaluate the economic impact of the long-term treatment benefits.
Article
Full-text available
A large proportion of older adults are affected by impaired glucose metabolism. Previous studies with fish protein have reported improved glucose regulation in healthy adults, but the evidence in older adults is limited. Therefore, we wanted to assess the effect of increasing doses of a cod protein hydrolysate (CPH) on postprandial glucose metabolism in older adults. The study was a double-blind cross-over trial. Participants received four different doses (10, 20, 30 or 40 mg/kg body weight (BW)) of CPH daily for 1 week with 1-week washout periods in between. The primary outcome was postprandial response in glucose metabolism, measured by samples of serum glucose and insulin in 20 min intervals for 120 min. The secondary outcome was postprandial response in plasma glucagon-like peptide 1 (GLP-1). Thirty-one subjects aged 60–78 years were included in the study. In a mixed-model statistical analysis, no differences in estimated maximum value of glucose, insulin or GLP-1 were observed when comparing the lowest dose of CPH (10 mg/kg BW) with the higher doses (20, 30 or 40 mg/kg BW). The estimated maximum value of glucose was on average 0·28 mmol/l lower when the participants were given 40 mg/kg BW CPH compared with 10 mg/kg BW ( P = 0·13). The estimated maximum value of insulin was on average 5·14 mIU/l lower with 40 mg/kg BW of CPH compared with 10 mg/kg BW ( P = 0·20). Our findings suggest that serum glucose and insulin levels tend to decrease with increasing amounts of CPH. Due to preliminary findings, the results require further investigation.
Article
Full-text available
Controlling of postprandial hyperglycemia using α-glucosidase inhibitors is one of the therapeutic approaches for the treatment of type 2 diabetes. Collagen hydrolysate, as an α-glucosidase inhibitor has not been reported. Therefore, This study intends to evaluate the activity and kinetics of inhibition of collagen hydrolysate against the α-glucosidase enzyme. Collagen hydrolysate was prepared from Thunnus albacares bone enzymatically using bacterial collagenase at hydrolysis times (0; 0.5; 1; 2; 3; 4; 5 and 6 h). The type of inhibition was determined by enzyme kinetics analysis. Collagen hydrolysate obtained at hydrolysis time of 1 h has the highest inhibitory activity of 24.47 %. Based on enzyme kinetics analysis, collagen hydrolysate showed a show a type of competitive inhibition. These results revealed that collagen hydrolysate has the potential as a natural antidiabetic in controlling blood glucose levels in type 2 diabetes.
Article
Full-text available
The term metabolic/cardiometabolic/insulin resistance syndrome could generally be defined as the co-occurrence of several risk factors inclusive of systemic arterial hypertension. Not only that organizations, such as the world health organization (WHO) have identified high blood pressure as one of the main risk factors of the cardiometabolic syndrome, but there is also a link between the occurrence of insulin resistance/impaired glucose tolerance and hypertension that would consequently lead to type-2 diabetes (T2D). Hypertension is medicated by various classes of synthetic drugs; however, severe or mild adverse effects have been repeatedly reported. To avoid and reduce these adverse effects, natural alternatives, such as bioactive peptides derived from different sources have drawn the attention of researchers. Among all types of biologically active peptides inclusive of marine-derived ones, this paper’s focus would solely be on fish and fishery by-processes’ extracted peptides and products. Isolation and fractionation processes of these products alongside their structural, compositional and digestion stability characteristics have likewise been briefly discussed to better address the structure-activity relationship, expanding the reader’s knowledge on research and discovery trend of fish antihypertensive biopeptides. Furthermore, drug-likeness of selected biopeptides was predicted by Lipinski’s rules to differentiate a drug-like biopeptide from nondrug-like one.
Article
Full-text available
Disturbances in glucose and lipid homeostasis are cardinal features of the metabolic syndrome that affect millions of people worldwide. These conditions have multi-organ impact, and while cardiovascular effects are usually the core for studies and preventive measures, other systems may also be affected, including the pancreas. Acute pancreatitis related to severe hypertriglyceridemia is an under-recognized condition that could lead to significant morbidity and mortality. Therefore, when suspected, prompt diagnosis and treatment should be initiated to cover the various aspects of this disorder. Though commonly known to be associated with excess of alcohol use, hypertriglyceridemia-related pancreatitis is particularly observed in diabetics, especially when uncontrolled. Here, we portray the possible mechanisms and clinical features that link type 2 diabetes, hypertriglyceridemia and pancreatitis, and discuss their health-related outcomes and the current and novel treatment options for this unique disease.
Article
Full-text available
Low-fat diets and energy restriction are recommended to prevent obesity and to induce weight loss, but high-protein diets are popular alternatives. However, the importance of the protein source in obesity prevention and weight loss is unclear. The aim of this study was to investigate the ability of different animal protein sources to prevent or reverse obesity by using lean or obese C57BL/6J mice fed high-fat/high-protein or low-fat diets with casein, cod or pork as protein sources. Only the high-fat/high-protein casein-based diet completely prevented obesity development when fed to lean mice. In obese mice, ad libitum intake of a casein-based high-fat/high-protein diet modestly reduced body mass, whereas a pork-based high-fat/high-protein diet aggravated the obese state and reduced lean body mass. Caloric restriction of obese mice fed high-fat/high-protein diets reduced body weight and fat mass and improved glucose tolerance and insulin sensitivity, irrespective of the protein source. Finally, in obese mice, ad libitum intake of a low-fat diet stabilized body weight, reduced fat mass and increased lean body mass, with the highest loss of fat mass found in mice fed the casein-based diet. Combined with caloric restriction, the casein-based low-fat diet resulted in the highest loss of fat mass. Overall, the dietary protein source has greater impact in obesity prevention than obesity reversal.
Article
Of many metabolic irregularity risk factors, glucose intolerance, insulin resistance, and abdominal obesity are the main ones. The risk of vascular diseases and cancers is rather high in such individuals therefore its treatment and prevention are equally important for the health of population. Marine including fish biopeptides have frequently shown to prevent and or ameliorate these risk factors. In our earlier studies, Atlantic mackerel hydrolysate and its fractions showed promising effects on immunity and hypertension. We, therefore, in the current study, aimed to examine the effects of mackerel hydrolysate along its cationic peptides, isolated at pH3 using electrodialysis with ultrafiltration membrane, on metabolic disorder factors in diet-induced insulin resistant obese mice. Mice were fed the chow and or the hypercaloric diet (high fat high sucrose) for the duration of 8-weeks. Subsequently the hypercaloric fed insulin resistant obese mice were gavaged daily either the vehicle and or the test material at the dosage of 208 mg of sample protein per kg mice body weight. According to our observations, mackerel peptides could not effectively modulate food intake, weight gain, adiposity, plasma glucose and insulin concentrations, hepatic cholesterol, and triglyceride levels. Thus, obesity and metabolic impairments could not be averted by the treatments in insulin resistant obese mice. In conclusion, anti-hypertensive, and immunomodulatory attributes of mackerel peptides at the tested concentrations do not seem to be correlated with its metabolic syndrome benefiting effects. To presume a general statement about the metabolic syndrome benefiting potency of Atlantic mackerel's biopeptides further research is warranted.