ArticlePDF AvailableLiterature Review

ANGPTL8 (Betatrophin) Role in Diabetes and Metabolic Diseases

Authors:

Abstract and Figures

Diabetes is a major disease worldwide that is reaching epidemic stage. Its increased prevalence as well as its association with a high number of complications such as cardiovascular diseases, nephropathy and retinopathy makes it an important disease for investigation. ANGPTL8 is a recently identified hormone that has been associated with two functionally important processes in the development of type 2 diabetes, insulin resistance as well as lipid metabolism. Initial work has showed that ANGPTL8 was expressed in liver, white adipose and brown adipose tissues. ANGPTL8 regulates the activity of lipoprotein lipase, which is a key enzyme in lipoprotein lipolysis pathway through its direct interaction with ANGPTL3. It has been also reported that it regulates the replication of β-cells in response to insulin resistance. As a result, many recent studies have focused on the association of ANGPTL8 with diabetes and obesity as well as its association with various metabolic markers in order to better understand its physiological role in glucose homeostasis and lipid metabolism. In this review we will highlight some of the key clinical findings, mainly from human studies, that investigated the role of ANGPTL8 in metabolic diseases such as diabetes, obesity and the metabolic syndrome.
Content may be subject to copyright.
This article has been accepted for publication and undergone full peer review but has not
been through the copyediting, typesetting, pagination and proofreading process which may
lead to differences between this version and the Version of Record. Please cite this article as
doi: 10.1002/dmrr.2919
This article is protected by copyright. All rights reserved.
ANGPTL8 (Betatrophin) Role in Diabetes and Metabolic
Diseases
Mohamed Abu-Farha1*, Jehad Abubaker1, Jaakko Tuomilehto2
1Biochemistry and Molecular Biology Unit; Dasman Diabetes Institute, Kuwait City, Kuwait. Dasman
2Diabetes Institute, Kuwait City, Kuwait.
*For correspondence:
Mohamed Abufarha, PhD, Biochemistry & Molecular Biology Unit
P.O. Box 1180, Dasman 15462, Kuwait
Phone: +965 2224 2999 Ext. 3010
mohamed.abufarha@dasmaninstitute.org; mafarha@gmail.com
This article is protected by copyright. All rights reserved.
Abstract
Diabetes is a major disease worldwide that is reaching epidemic stage. Its increased
prevalence as well as its association with a high number of complications such as
cardiovascular diseases, nephropathy and retinopathy makes it an important disease for
investigation. ANGPTL8 is a recently identified hormone that has been associated with two
functionally important processes in the development of type 2 diabetes, insulin resistance as
well as lipid metabolism. Initial work has showed that ANGPTL8 was expressed in liver,
white adipose and brown adipose tissues. ANGPTL8 regulates the activity of lipoprotein
lipase, which is a key enzyme in lipoprotein lipolysis pathway through its direct interaction
with ANGPTL3. It has been also reported that it regulates the replication of β-cells in
response to insulin resistance. As a result, many recent studies have focused on the
association of ANGPTL8 with diabetes and obesity as well as its association with various
metabolic markers in order to better understand its physiological role in glucose homeostasis
and lipid metabolism. In this review we will highlight some of the key clinical findings,
mainly from human studies, that investigated the role of ANGPTL8 in metabolic diseases
such as diabetes, obesity and the metabolic syndrome.
This article is protected by copyright. All rights reserved.
INTRODUCTION
Diabetes is reaching an epidemic stage worldwide with International Diabetes
Federation (IDF) estimating that around 415 million adults are diabetic and this number will
increase to approximately 640 million by the year 2040 [1]. Since hyperglycaemia increases
progressively during the development of diabetes, there is a high number of people with pre-
diabetes in a population at any given time. Approximately half of these people will become
diabetic within 10 years, if no preventive intervention is provided [2]. Generally, diabetes
develops from the inability of the pancreas to cope with the increased insulin demand in type
2 diabetes (T2D) [3-7] or the inability of β -cells to produce insulin due to a destruction of β-
cells in type 1 diabetes (T1D) [8-10]. Adequate glycaemic control in diabetic patients can be
achieved through a number of approaches such as lifestyle interventions and various glucose
lowering drugs including insulin injections [11, 12] [13]. In T2D the beta-cell inability to
cope with the increased insulin demand can be caused by multiple factors including genetic
factors, aging, obesity as well as oxidative stress. Traditionally, T2D has been diagnosed as
elevated fasting blood glucose (FBG) or post-challenge glucose after an oral glucose
tolerance test (OGTT) [14], but recently also glycated haemoglobin A1c (HbA1c) has been
introduced as another measure to diagnose diabetes [15]. Since various pharmacologic and
non-pharmacologic management options are currently available, early detection of diabetes is
crucial to achieve proper control of diabetes and its complications.
Active screening for diabetes can be carried out starting with non-laboratory diabetes
risk scores that have been developed and validated in various populations. Another approach
is to identify biomarkers that predict the future development of diabetes, its severity as well
as the severity of its complications. These may offer great advantages to delay or prevent the
onset of diabetes or its complications. In this review we will focus on a recently discovered
biomarker called ANGPTL8 or betatrophin that has been shown to play a key role in lipid
metabolism as well as β-cell proliferation [16-19]. ANGPTL8 is the name that will be mostly
used in this paper. This biomarker has also been suggested to be an independent predictor of
the development of T2D [20].
IDENTIFICATION OF ANGPTL8
One of the recent biomarkers that has been recognized to play a role in glucose as
well as lipid homeostasis is ANGPTL8 [19-24]. This protein is an atypical ANGPTL protein
This article is protected by copyright. All rights reserved.
as it lacks the fibrinogen-like domain and only possesses the N-terminal coiled-coil domains
[21, 24]. This protein has been given many names, in addition to ANGPTL8, it has been
called hepatocellular carcinoma-associated gene (TD26) as well as refeeding induced in
fat and liver (RIFL) [22] and lipasin [24, 25]. RIFIL and lipasin names were given based
on its function in fasting/feeding regulation as well as lipoprotein lipase (LPL) activity
regulation respectively [22], [24, 25]. Initial work in mice has shown that mice lacking the
ANGPTL8 (or Gm6484, as referred to in mice) had low triglyceride (TG) level in blood
circulation [26]. This gene was earlier named RIFL by Ren et al. where they showed that it
was mainly expressed in white and brown adipose tissues (WAT and BAT, respectively) as
well as liver tissues and was regulated by nutrients and hormonal factors [22]. They showed
that TG level in ANGPTL8-null mice was only one third of that found in the wild type.
Knocking down RIFL in 3T3-L1 cells resulted in around 35% reduction in TG concentration.
ANGPTL8 expression was increased over 100 folds during 3T3-L1 cells adipogenesis.
Furthermore, ANGPTL8 expression was increased by about eight times in WAT of ob/ob
mice (an obesity mouse model) compared to wild type mice. Moreover, both WAT and liver
RIFL gene expression was increased by about 100 and 12 times, respectively, after feeding
and fasting conditions in mice. RIFL was induced by insulin and suppressed by forskolin,
isoproterenol and dibutyryl-cAMP in 3T3-L1 cells. Taken together, the data by Ren et al.
confirmed the involvement of RIFL in adipogenesis and lipid metabolism.
Concurrently, a second research group also showed that ANGPTL8 or lipasin, as they
referred to it, was capable of inhibiting LPL activity in vitro [23]. Overexpressing ANGPTL8
in mice lead to about a 5-fold increase in plasma TG. Furthermore, ANGPTL8 expression
was increased in liver during a high fat diet and reduced after fasting. They also showed that
ANGPTL8 is thermo-modulated by cold as its expression was induced in BAT after cold
exposure [24]. It can be concluded from these previous studies that ANGPTL8 is mostly
expressed in liver, WAT and BAT [21-24]. The level of ANGPTL8 can be induced by high
fat feeding and exposure to cold. Also, ANGPTL8 is involved in regulating plasma TG level
through its inhibition of LPL activity, which results from its interaction with the N-terminal
domain of ANGPTL3 [21-24].
A third critical paper was published by Quagliarini et el. also in 2012 showing similar
tissue distribution with particular enrichment in liver and adipose tissue [21]. ANGPTL8 was
This article is protected by copyright. All rights reserved.
given this name based on its sequence similarity to members of the angiopoietin-like protein
family mainly ANGPTL3, which was also recognized as an interaction partner for
ANGPTL8. Adding further support to ANGPTL8 role in lipid metabolism, Quagliarini et el.
showed that a non-synonymous SNP (R59W) was associated with lower LDL-C and HDL-C.
Using the Dallas Heart Study (DHS), this variant was associated with lower plasma levels of
LDL-C and HDL-C in African Americans and Hispanics but not European Americans [21].
Their results were replicated in African Americans in the Atherosclerosis Risk in
Communities (ARIC) study [21]. No association was observed between this variant and TG
level in any ethnic group [21]. We have recently shown that people of Arab origin who
carried this variant had higher FBG [27]. In addition, Quagliarini et al. showed that the
overexpression of ANGPTL8 lead to increased TG level that was further increased in the
presence of ANGPTL3 [21]. This suggested that the two proteins maybe acting in synergy to
control TG level. Indeed, this was validated when they showed that ANGPTL8 protein was
physically interacting with ANGPTL3, which resulted in regulating the plasma lipoprotein
level. They also showed that overexpressing ANGPTL3 alone in wild type mice did not result
in increased TG while it was true for the overexpression of ANGPTL8. Collectively, their
findings suggested that ANGPTL8 plays a role in regulating the level of TG through its
interaction with ANGPTL3 forming a complex that might act as a LPL inhibitor.
Subsequent publications showed that ANGPTL8 was involved in β-cell proliferation
in response to treatment with insulin receptor antagonist (S961). In a study by Yi et al. it was
reported that a 17 fold increase in β-cell proliferation was observed upon overexpression of
ANGPTL8 betatrophin in mouse liver, leading to a 3-fold increase in β-cell mass. They
also showed that ANGPTL8 was released into circulation, and then binds to β-cells through
an unidentified receptor leading to increased β-cell proliferation and mass [19]. Furthermore,
they reported that mice overexpressing ANGPTL8 had improved glucose tolerance and lower
FBG [19]. These conclusions were however questioned by other studies that showed that
mice lacking ANGPTL8 had normal glucose and insulin tolerance [28]. Furthermore, one
study showed that ANGPTL8 did not have any effect on β-cell expansion in mice and further
confirmed that blood TG was reduced by ANGPTL8 deletion and increased after its
overexpression [29]. The only consistent finding reported by different studies is that showing
that knocking down ANGPTL8 expression in mice results in disrupted TG delivery to
adipose tissues [21, 28]. Another study by Jiao et al. showed that transplantation of human
and mice islets treated with S961 under the kidney capsule in mice resulted in increased
This article is protected by copyright. All rights reserved.
ANGPTL8 level, which in turn caused an increase in beta-cell proliferation in the
transplanted mice islets [30]. However, this effect was not observed in human transplanted
islets. In conclusion, the role of ANGPTL8 in beta-cell proliferation in humans as well as
mice has been challenged, however, its role in insulin resistance is becoming well established
even though more studies investigating its mechanism of action remain to be conducted [29,
31]. This review will focus more on the clinical studies that looked at the plasma level of
ANGPTL8 and its possible association with lipid and glucose profiles as well as other
diabetes and obesity related biomarkers.
ANGPTL8 ROLE IN OBESITY
Obesity is the major risk factor for T2D that leads to increased insulin demand and
insulin resistance. In combination with lack of physical activity and genetic predisposition,
obesity will stress beta-cells to increase insulin production reaching a point where beta-cells
fail to compensate for the increased insulin demand leading to permanently elevated glucose
concentration in blood circulation, i.e. the development of T2D [13]. ANGPTL8 is produced
by two of the key tissues in the insulin resistance pathways, liver and adipose tissue. Liver is
one of the main tissues involved in glucose metabolism. Adipose tissues also plays a major
role in insulin resistance through the increased inflammation, free fatty acids release, and
reduced adiponectin production [32, 33]. Given the connection between obesity and insulin
resistance as well as lipid metabolism, it’s imperative that ANGPTL8 would be regulated by
obesity.
Studies in rodents has shown that ANGPTL8 level was increased in obesity [21-23].
We have recently published a large human cohort study that showed a positive association
between ANGPTL8 and BMI, and also with waist/hip ratio in non-diabetic people [20]. In
another study we also showed that ANGPTL8 was increased in obese people, but its level
was reduced after exercise [34]. In this study we looked at both forms of ANGPTL8 which
has been called the C-terminal 139-198 and the full length form of ANGPTL8 due to the
different ELISA antibodies used to measure its plasma level. The full length level of
ANGPTL8 was measured using ELISA (Wuhan EIAAB Science co) (catalogue number
E1164H) as described previously. Whereas, the C-terminal 139-198 form was measured
using ELISA kit recognizing the C-terminal domain of ANGPTL8 from 139 to 198 amino
acid manufactured by Phoenix Pharmaceuticals (catalogue number EK-051-55). The use of
This article is protected by copyright. All rights reserved.
multiple ELISA kits has been suggested as the cause of some of the discrepancies in the
reported data regarding ANGPTL8 level under various conditions such as obesity as well as
its association with FBG [35]. The accuracy of both full length and C-terminal 139-198
ELISA kits has been evaluated by Fu et al. showing that the correlation coefficient between
both kits was 0.99. It was also shown that the C-terminal kit gave higher values for
ANGPTL8 plasma level when compared with the levels obtained from the full length form.
This finding suggested to result from the possibility that the full length ANGPTL8 might be
degraded, therefore its level will be less than those obtained for the C-terminal 139-198 form
[35]. We have shown that both forms were increased in obese people. However, only the full
length ANGPTL8 form was positively associated with FBG [34]. Similarly, other studies
reported increased ANGPTL8 level in obesity as shown in Table 1. For example, Fu et al.
showed that ANGPTL8 level was positively correlated with BMI (rho=0.49, P<0.001) [36].
Taken together, our data suggest that kit variations can explain the differential association
between ANGPTL8 and FBG but not the changes in obesity level as both kits showed a
similar trend. Ethnic variations or other unmeasured factors might be responsible for some of
these changes.
Guo et al. showed that ANGPTL8 was increased in overweight people compared
with lean controls [37], but obese people, on the other hand, had a lower ANGPTL8 level
than overweight people [37]. Moreover, in another study by Fenzl et al. the ANGPTL8 level
between lean and morbid obese people was compared, but they did not observe a significant
difference between the groups [38]. Only one study has reported reduced ANGPTL8 level in
obesity; ANGPTL8 level was reduced in obesity and even more so in obese people with
insulin resistance [39]. One of the main differences observed with this study was the use of a
different kit, which was purchased from CUSABIO/ Aviscera Bioscience, than what has been
used in most other studies. Their ANGPTl8 levels were ranging from 45.1-8.8 ng/mL, which
is roughly 10-40 times higher than levels observed by other kits [39].
The varying results observed in the reported ANGPTL8 levels may also be in part due
to other comorbidities that can play a role in regulating the ANGPTL8 levels. For example it
has been reported that ANGPTL8 level may be affected by thyroid function [40], polycystic
ovary syndrome [41] as well as metabolic syndrome (MetS) [42, 43]. We have also recently
shown that ANGPTL8 was higher in people with MetS than those without. When stratified
according to the number of MetS components, people with higher number of MetS
components had significantly higher level of ANGPTL8 [44]. Similarly, Crujeiras et al. have
This article is protected by copyright. All rights reserved.
recently shown that ANGPTL8 was increased in obese people with MetS and it was
associated with less reduction in adiposity and dyslipidemia after a weight-loss program [42].
ANGPTL8 ROLE IN DIABETES
T2D develops as a result of the failure of beta-cells to compensate for the increased
insulin demand caused by hyperglycemia and insulin resistance [45-48]. The search for
agents that promote beta-cell proliferation has been very active yet elusive [17, 18]. The
discovery of ANGPTL8 as a hormone that is capable of increasing beta-cell proliferation has
been hailed as a scientific breakthrough [16-18]. It has been shown that ANGPTL8 was
increased under states of insulin resistance to increase beta-cell proliferation and increase
insulin production [19]. ANGPTL8 was increased in ob/ob mice and in the db/db diabetic
mouse model [19]. These findings, particularly the ability of ANGPTL8 to increase beta-cell
proliferation was later challenged [29, 49, 50]. Similarly, many studies have shown that
levels of ANGTPL8 was increased in people with T2D subjects as summarized in Table 1.
This is contrary to its proposed role in increasing beta-cell proliferation where a decrease in
its level is expected. It can be speculated that a state of ANGPTL8 resistance is observed in
these people with T2D where beta-cells are not responsive to the increased ANGPTL8 level.
We have recently published the largest cohort study showing that ANGPTL8 was
increased in people with T2D [20]. We compared the level of ANGPTL8 in 556 people with
T2D with that of 1047 non-diabetic people: ANGPTL8 level was about three times higher in
people with T2D. We also showed that ANGPTL8 was an independent predictor of T2D.
People in the highest tertiles of ANGPTL8 had about 6-fold risk of developing T2D after
adjusting for multiple factors such as age, gender, ethnicity and lipid profile. In our
population, ANGPTL8 was positively associated with many factors such as age, BMI,
waist/hip ratio, FBG, HbA1c, HOMA-IR in non-diabetic people. Our results suggested that
the increased ANGPTL8 level in people with T2D was not effective in increasing insulin
production and was not affecting plasma glucose level [20]. This conclusion was further
supported by the finding that ANGPTL8 was not associated with C-peptide level in the
people with T2D unlike the non-diabetic people in whom ANGPTL8 was positively
associated with the C-peptide level [51].
Other studies have also shown that ANGPTL8 was increased in people with T2D [36,
52-55]. Espes et al. showed that plasma ANGPTL8 was increased in 27 people with T2D vs.
This article is protected by copyright. All rights reserved.
18 gender, age, and BMI matched controls [52]. In an earlier study they also looked at the
changes in ANGPTL8 level in people with T1D and showed that ANGPTL8 was increased
among them [56]. However, this increase in ANGPTL8 level was not associated with any
increase in residual C-peptide level. Fu et al. showed that serum ANGPTL8 (or lipasin as
they referred to it) was increased in 14 diabetic people compared with 15 BMI and age
matched non-diabetic people, and it was correlated with plasma glucose [36]. Hu et al.
looked at ANGPTL8 in 83 newly diagnosed people with T2D and 83 age, sex and BMI
matched healthy controls; ANGPTL8 level was about two times higher in people with T2D,
and that ANGPTL8 was positively associated with post-challenge serum 2-h OGTT and
insulin [53]. Similarly, Chen et al. measured ANGPTL8 in a subset of 330 individuals from
the Risk Evaluation of Cancers in Chinese Diabetic Individuals. They were divided into four
groups matched for age, sex, BMI, and lipid profile. The group 1 comprised 137 people with
normal glucose tolerance, the group 2 69 people with isolated impaired FBG, the group 3 120
people with isolated impaired glucose tolerance and the 4th group 112 people with newly
diagnosed T2D. The ANGPTL8 level in people with T2D was the highest amongst all groups
and associated with insulin resistance indices [54]. A Japanese study comprising 12 healthy
controls, 34 people with T1D and 30 people with T2D showed that ANGPTL8 was higher in
both T1D and T2D subjects than in non-diabetic controls [55]. On the other hand, some
studies have found ANGPTL8 level decreased in people with T2D. Gomez-Ambrosi et al.
studied 153 people (75 obese normo-glycemic, 30 obese with impaired glucose tolerance and
15 obese with T2D) matched for sex, age and body adiposity and compared to 33 normo-
glycemic lean subjects. However, as highlighted earlier, they used different ELISA kit to
measure the plasma level of ANGPTL8 [39]. Their data showed that ANGPTL8 was
decreased in obese people and further decreased in people with T2D subjects. They also
showed that ANGPTL8 was positively associated with the insulin sensitivity as measured by
QUICKI index (r=0.46, P<0.001). In another study, also using the same ELISA kit
manufactured by CUSABIO/ Aviscera Bioscience, it was shown that ANGPTL8 was
increased in overweight people with T2D compared to their lean counterparts
[37];ANGPTL8 was not significantly different between the people with T2D and the non-
diabetic people in their population, andANGPTL8 was positively correlated with insulin and
insulin resistance as measured by HOMA-IR. One of the main reasons for the different
results reported by these two groups [39, 57] compared with other researchers is likely the
different ELISA kit they used.
This article is protected by copyright. All rights reserved.
A recent meta-analysis has investigated the association between ANGPTL8 level and
T2D through searching PubMed and Embase for studies published comparing ANGPTL8
level between people with and without T2D; nine studies met their criteria and were included
in this meta-analysis [58]. The level of ANGPTL8 was found higher in people with T2D and
that ANGPTL8 level was higher in non-obese people with T2D than non-diabetic non-obese
people. On the other hand, ANGPTL8 was not significantly different in obese people with
T2D compared to obese non-diabetic people. It is important to highlight that our more recent
data, where we showed that ANGPTL8 level was higher in people with T2D, was not
included in their meta-analysis. Our study included a larger sample than these previous
smaller studies. A small sample size is always an important factor in the interpretation of
results from meta-analyses, and it was correctly mentioned by Li et al. as one of the main
limitations of the studies in their meta-analysis [58]. Another limitation was the use of
variable ELISA kits to measure the level of ANGPTL8 and the wide variation in actual
ANGPTL8 levels reported among different studies [58].
In addition to its increased level in people with T2D, ANGPTL8 has been found to be
increased in women with gestational diabetes (GDM) either in blood circulation [57, 59-61]
or cord blood [60, 61] as summarized in Table 2. Consistently, ANGPTL8 has been initially
shown to be increased during gestation in a mouse model [19]. Yi et al showed that
ANGPTL8 was almost 20 times higher in 18.5 dpc (date post-conception) mice compared
with non-pregnant female mice or mice in their early pregnancy [19]. The increased
ANGPTL8 level during pregnancy has been suggested to stem from a physiological
compensatory mechanism to increase beta-cell proliferation. During pregnancy females
experience a change in hormonal balance that results in a progressive decline in insulin
sensitivity to a level that imitates insulin resistance observed in T2D. The majority of females
do not exhibit changes in their blood glucose throughout pregnancy due to the adequate β-cell
compensation. In the situation where insulin resistance is stronger than what the β-cells can
compensate for, or the β-cell function declines, GDM may develop. It has been suggested that
increased ANGPTL8 level will promote beta-cell proliferation in order to compensate for the
increased insulin demand. In concordance with this argument, Ebert et al. showed that
ANGPTL8 level was higher in women with GDM compared with healthy pregnant women
[62]. Additionally, they showed that ANGPTL8 level was generally lowered postpartum from
its level during pregnancy. Similarly, Erol et al. found the ANGPTL8 level was significantly
higher in 45 pregnant women diagnosed with GDM compared with 45 healthy pregnant
This article is protected by copyright. All rights reserved.
women matched for BMI and gestational age [59]. Other studies have shown that ANGPTL8
level was increased in the cord blood from women with GDM compared with healthy women
at delivery [57, 60, 61].
ANGTPL8 AND ITS INTERACTION WITH OTHER BIOMARKERS
In addition to its association with ANGPTL3 [21, 28] and insulin [19, 20, 22], the
association of ANGPTL8 with other metabolic markers such as high sensitivity C-reactive
protein (hsCRP), C-Peptide and irisin has been investigated [42, 51, 52, 56, 57, 60, 62, 63].
We found an association between ANGPTL8 and C-peptide in an attempt to understand
whether the increase in ANGPTL8 production will have an impact on the endogenous insulin
production as measured by plasma C-peptide. Comparing ANGPTL8 level in 535 non-
diabetic and 214 T2D people ANGPTL8 was significantly associated with C-peptide level in
the non-diabetic group only [51]. The lack of such association in the T2D group suggested
that the increased ANGPTL8 level observed in people with T2D was not due to increased C-
peptide production [51]. Espes et al. showed that increased ANGPTL8 in people with T1D
was not associated with an increased C-peptide level [56]. They also showed that ANGPTL8
did not correlate with C-peptide in either non-diabetic or T2D people in a small study [52].
On the other hand, Trebotic et al. showed an inverse correlation between ANGPTL8 and C-
peptide in pregnant women, both in non-diabetic pregnant women and those with GDM [57].
ANGPTL8 expression in brown adipose tissues has been recently linked to the action
of another browning related myokine called irisin [64]. In cell models, irisin has been found
to increase the expression of ANGPTL8 during adipocyte differentiation. It has been shown
that ANGPTL8 and irisin were positively correlated with ANGPTL8 level in people with
T1D [65], and that the level of irisin was a positive and independent predictor of the post-
delivery ANGPTL8 level [66]. In another study of 120 people with T2D, ANGPTL8 was not
shown to be associated with irisin in T2D people [67]. Taken together, these inconsistent
findings between ANGPTL8 and irisin highlight the need for more studies to address this
issue.
Based on its role in lipid regulation, ANGPTL8 has been also suggested as a potential
therapeutic target for dyslipidemia [21, 68]. In addition, since the ANGPTL8 level has been
shown to be increased in obesity and in people with MetS [43] we evaluated the potential
association between ANGPTL8 and low-grade inflammation and found a positive association
This article is protected by copyright. All rights reserved.
between ANGPTL8 and hsCRP [43]. A positive association between ANGPTL8 and
hsCRP has been also found in women diagnosed with polycystic ovary syndrome [69].
CONCLUSION
In conclusion, markers that can detect the development of diabetes can greatly
enhance our understanding of the pathophysiology of the disease and may be used to suggest
methods for prevention. ANGPTL8 is a promising biomarker for early detection of T2D as
well as for GDM in pregnant women. Most studies have shown that ANGPTL8 is increased
in people with T2D, T1D and GDM. It is also increased in obese people and those with MetS.
Nonetheless, some discrepancies in the data exist, most likely due to small sample sizes in the
early studies and due to the wide range of detection kits used to assay the levels ANGPTL8
without any standardization among them. Also, since most studies to date are cross sectional,
the exact role of ANGPTL8 in the pathophysiology of diabetes is not yet very clear and its
contribution to beta-cell proliferation and insulin resistance is far from being well understood.
Taken together, ANGPTL8 is a protein that with more novel studies may significantly
advance our understanding of the pathophysiology of the development of diabetes and other
metabolic diseases.
ACKNOWLEDGMENT
We are indebted to Kuwait Foundation for the Advancement of Sciences (KFAS) for
financial support of by the funding provided to Dasman Diabetes Institute through project
numbers (RA-2014-021 and ACC-14013003). The funding agency was not involved in any
aspect pertinent to this article. None of the authors have been paid to write this article by a
pharmaceutical company or other agency. We are also thankful to Ms. Irina Al-khairi for her
efforts in proofreading this article. None of the authors (MA, JA and JT) have any conflict of
interest or anything to disclose.
Author Contributions
MA: Wrote the manuscript. JA: Critically revised the manuscript. JT: Critically revised
manuscript.
This article is protected by copyright. All rights reserved.
REFERENCES
1. Federation ID: IDF diabetes atlas: 7th edition http://wwwdiabetesatlasorg/ 2015.
2. Lindstrom J, Ilanne-Parikka P, Peltonen M, Aunola S, Eriksson JG, Hemio K, Hamalainen H,
Harkonen P, Keinanen-Kiukaanniemi S, Laakso M, et al: Sustained reduction in the incidence
of type 2 diabetes by lifestyle intervention: follow-up of the Finnish Diabetes Prevention
Study. Lancet 2006, 368:1673-1679.
3. Rhodes CJ, White MF, Leahy JL, Kahn SE: Direct autocrine action of insulin on beta-cells:
does it make physiological sense? Diabetes 2013, 62:2157-2163.
4. Johnson AM, Olefsky JM: The origins and drivers of insulin resistance. Cell 2013, 152:673-
684.
5. Drucker DJ: Incretin action in the pancreas: potential promise, possible perils, and
pathological pitfalls. Diabetes 2013, 62:3316-3323.
6. Ashcroft FM, Rorsman P: Diabetes mellitus and the beta cell: the last ten years. Cell 2012,
148:1160-1171.
7. Marchetti P, Bugliani M, Boggi U, Masini M, Marselli L: The pancreatic beta cells in human
type 2 diabetes. Adv Exp Med Biol 2012, 771:288-309.
8. Skog O, Korsgren S, Melhus A, Korsgren O: Revisiting the notion of type 1 diabetes being a
T-cell-mediated autoimmune disease. Curr Opin Endocrinol Diabetes Obes 2013, 20:118-
123.
9. Huber A, Menconi F, Corathers S, Jacobson EM, Tomer Y: Joint genetic susceptibility to type
1 diabetes and autoimmune thyroiditis: from epidemiology to mechanisms. Endocr Rev
2008, 29:697-725.
10. Bluestone JA, Tang Q, Sedwick CE: T regulatory cells in autoimmune diabetes: past
challenges, future prospects. J Clin Immunol 2008, 28:677-684.
11. Tuomilehto J, Lindstrom J, Eriksson JG, Valle TT, Hamalainen H, Ilanne-Parikka P, Keinanen-
Kiukaanniemi S, Laakso M, Louheranta A, Rastas M, et al: Prevention of type 2 diabetes
mellitus by changes in lifestyle among subjects with impaired glucose tolerance. N Engl J
Med 2001, 344:1343-1350.
12. Type 2 diabetes mellitus. Nature Reviews Disease Primers 2015, 1:15039.
13. DeFronzo RA, Ferrannini E, Groop L, Henry RR, Herman WH, Holst JJ, Hu FB, Kahn CR, Raz I,
Shulman GI, et al: Type 2 diabetes mellitus. Nat Rev Dis Primers 2015, 1:15019.
14. Diabetes mellitus. Report of a WHO Study Group. World Health Organ Tech Rep Ser 1985,
727:1-113.
15. International Expert C: International Expert Committee report on the role of the A1C assay
in the diagnosis of diabetes. Diabetes Care 2009, 32:1327-1334.
16. Crunkhorn S: Metabolic disorders: Betatrophin boosts beta-cells. Nat Rev Drug Discov 2013,
12:504.
17. Kugelberg E: Diabetes: Betatrophin--inducing beta-cell expansion to treat diabetes
mellitus? Nat Rev Endocrinol 2013, 9:379.
18. Lickert H: Betatrophin fuels beta cell proliferation: first step toward regenerative therapy?
Cell Metab 2013, 18:5-6.
19. Yi P, Park JS, Melton DA: Betatrophin: a hormone that controls pancreatic beta cell
proliferation. Cell 2013, 153:747-758.
20. Abu-Farha M, Abubaker J, Al-Khairi I, Cherian P, Noronha F, Hu FB, Behbehani K, Elkum N:
Higher plasma betatrophin/ANGPTL8 level in Type 2 Diabetes subjects does not correlate
with blood glucose or insulin resistance. Sci Rep 2015, 5:10949.
21. Quagliarini F, Wang Y, Kozlitina J, Grishin NV, Hyde R, Boerwinkle E, Valenzuela DM, Murphy
AJ, Cohen JC, Hobbs HH: Atypical angiopoietin-like protein that regulates ANGPTL3. Proc
Natl Acad Sci U S A 2012, 109:19751-19756.
This article is protected by copyright. All rights reserved.
22. Ren G, Kim JY, Smas CM: Identification of RIFL, a novel adipocyte-enriched insulin target
gene with a role in lipid metabolism. Am J Physiol Endocrinol Metab 2012, 303:E334-351.
23. Zhang R: Lipasin, a novel nutritionally-regulated liver-enriched factor that regulates serum
triglyceride levels. Biochem Biophys Res Commun 2012, 424:786-792.
24. Fu Z, Yao F, Abou-Samra AB, Zhang R: Lipasin, thermoregulated in brown fat, is a novel but
atypical member of the angiopoietin-like protein family. Biochem Biophys Res Commun
2013, 430:1126-1131.
25. Zhang R, Abou-Samra AB: Emerging roles of Lipasin as a critical lipid regulator. Biochem
Biophys Res Commun 2013, 432:401-405.
26. Tang T, Li L, Tang J, Li Y, Lin WY, Martin F, Grant D, Solloway M, Parker L, Ye W, et al: A
mouse knockout library for secreted and transmembrane proteins. Nat Biotechnol 2010,
28:749-755.
27. Abu-Farha M, Melhem M, Abubaker J, Behbehani K, Alsmadi O, Elkum N:
ANGPTL8/Betatrophin R59W variant is associated with higher glucose level in non-diabetic
Arabs living in Kuwaits. Lipids Health Dis 2016, 15:26.
28. Wang Y, Quagliarini F, Gusarova V, Gromada J, Valenzuela DM, Cohen JC, Hobbs HH: Mice
lacking ANGPTL8 (Betatrophin) manifest disrupted triglyceride metabolism without
impaired glucose homeostasis. Proc Natl Acad Sci U S A 2013, 110:16109-16114.
29. Gusarova V, Alexa CA, Na E, Stevis PE, Xin Y, Bonner-Weir S, Cohen JC, Hobbs HH, Murphy AJ,
Yancopoulos GD, Gromada J: ANGPTL8/betatrophin does not control pancreatic beta cell
expansion. Cell 2014, 159:691-696.
30. Jiao Y, Le Lay J, Yu M, Naji A, Kaestner KH: Elevated mouse hepatic betatrophin expression
does not increase human beta-cell replication in the transplant setting. Diabetes 2014,
63:1283-1288.
31. Stewart AF: Betatrophin versus bitter-trophin and the elephant in the room: time for a
new normal in beta-cell regeneration research. Diabetes 2014, 63:1198-1199.
32. Boden G, Shulman GI: Free fatty acids in obesity and type 2 diabetes: defining their role in
the development of insulin resistance and beta-cell dysfunction. Eur J Clin Invest 2002, 32
Suppl 3:14-23.
33. O'Rahilly S: Human obesity and insulin resistance: lessons from experiments of nature.
Biochem Soc Trans 2007, 35:33-36.
34. Abu-Farha M, Sriraman D, Cherian P, AlKhairi I, Elkum N, Behbehani K, Abubaker J:
Circulating ANGPTL8/Betatrophin Is Increased in Obesity and Reduced after Exercise
Training. PLoS One 2016, 11:e0147367.
35. Fu Z, Abou-Samra AB, Zhang R: An explanation for recent discrepancies in levels of human
circulating betatrophin. Diabetologia 2014.
36. Fu Z, Berhane F, Fite A, Seyoum B, Abou-Samra AB, Zhang R: Elevated circulating
lipasin/betatrophin in human type 2 diabetes and obesity. Sci Rep 2014, 4:5013.
37. Guo K, Lu J, Yu H, Zhao F, Pan P, Zhang L, Chen H, Bao Y, Jia W: Serum betatrophin
concentrations are significantly increased in overweight but not in obese or type 2 diabetic
individuals. Obesity (Silver Spring) 2015, 23:793-797.
38. Fenzl A, Itariu BK, Kosi L, Fritzer-Szekeres M, Kautzky-Willer A, Stulnig TM, Kiefer FW:
Circulating betatrophin correlates with atherogenic lipid profiles but not with glucose and
insulin levels in insulin-resistant individuals. Diabetologia 2014.
39. Gomez-Ambrosi J, Pascual E, Catalan V, Rodriguez A, Ramirez B, Silva C, Gil MJ, Salvador J,
Fruhbeck G: Circulating Betatrophin Concentrations Are Decreased in Human Obesity and
Type 2 Diabetes. J Clin Endocrinol Metab 2014:jc20141568.
40. Han C, Xia X, Liu A, Zhang X, Zhou M, Xiong C, Liu X, Sun J, Shi X, Shan Z, Teng W: Circulating
Betatrophin Is Increased in Patients with Overt and Subclinical Hypothyroidism. Biomed
Res Int 2016, 2016:5090852.
This article is protected by copyright. All rights reserved.
41. Erbag G, Eroglu M, Turkon H, Sen H, Binnetoglu E, Aylanc N, Asik M: Relationship between
betatrophin levels and metabolic parameters in patients with polycystic ovary syndrome.
Cell Mol Biol (Noisy-le-grand) 2016, 62:20-24.
42. Crujeiras AB, Zulet MA, Abete I, Amil M, Carreira MC, Martinez JA, Casanueva FF: Interplay
of atherogenic factors, protein intake and betatrophin levels in obese-metabolic syndrome
patients treated with hypocaloric diets. Int J Obes (Lond) 2016, 40:403-410.
43. Abu-Farha M, Abubaker J, Al-Khairi I, Cherian P, Noronha F, Kavalakatt S, Khadir A,
Behbehani K, Alarouj M, Bennakhi A, Elkum N: Circulating angiopoietin-like protein 8
(betatrophin) association with HsCRP and metabolic syndrome. Cardiovasc Diabetol 2016,
15:25.
44. Abu-Farha M, Abubaker J, Al-Khairi I, Cherian P, Noronha F, Kavalakatt S, Khadir A,
Behbehani K, Alarouj M, Bennakhi A, Elkum N: Circulating angiopoietin-like protein 8
(betatrophin) association with HsCRP and metabolic syndrome. Cardiovasc Diabetol 2016,
15:25.
45. Kassem SA, Ariel I, Thornton PS, Scheimberg I, Glaser B: Beta-cell proliferation and
apoptosis in the developing normal human pancreas and in hyperinsulinism of infancy.
Diabetes 2000, 49:1325-1333.
46. Bonner-Weir S, Weir GC: New sources of pancreatic beta-cells. Nat Biotechnol 2005, 23:857-
861.
47. Karaca M, Magnan C, Kargar C: Functional pancreatic beta-cell mass: involvement in type 2
diabetes and therapeutic intervention. Diabetes Metab 2009, 35:77-84.
48. Kulkarni RN, Mizrachi EB, Ocana AG, Stewart AF: Human beta-cell proliferation and
intracellular signaling: driving in the dark without a road map. Diabetes 2012, 61:2205-
2213.
49. Cox AR, Lam CJ, Bonnyman CW, Chavez J, Rios JS, Kushner JA: Angiopoietin-like protein 8
(ANGPTL8)/betatrophin overexpression does not increase beta cell proliferation in mice.
Diabetologia 2015.
50. Yi P, Park JS, Melton DA: Perspectives on the activities of ANGPTL8/betatrophin. Cell 2014,
159:467-468.
51. Abu-Farha M, Abubaker J, Noronha F, Al-Khairi I, Cherian P, Alarouj M, Bennakhi A, Elkum N:
Lack of associations between betatrophin/ANGPTL8 level and C-peptide in type 2 diabetic
subjects. Cardiovasc Diabetol 2015, 14:112.
52. Espes D, Martinell M, Carlsson PO: Increased circulating betatrophin concentrations in
patients with type 2 diabetes. Int J Endocrinol 2014, 2014:323407.
53. Hu H, Sun W, Yu S, Hong X, Qian W, Tang B, Wang D, Yang L, Wang J, Mao C, et al: Increased
Circulating Levels of Betatrophin in Newly Diagnosed Type 2 Diabetic Patients. Diabetes
Care 2014.
54. Chen X, Lu P, He W, Zhang J, Liu L, Yang Y, Liu Z, Xie J, Shao S, Du T, et al: Circulating
betatrophin levels are increased in patients with type 2 diabetes and associated with
insulin resistance. J Clin Endocrinol Metab 2015, 100:E96-100.
55. Yamada H, Saito T, Aoki A, Asano T, Yoshida M, Ikoma A, Kusaka I, Toyoshima H, Kakei M,
Ishikawa SE: Circulating betatrophin is elevated in patients with type 1 and type 2 diabetes.
Endocr J 2015.
56. Espes D, Lau J, Carlsson PO: Increased circulating levels of betatrophin in individuals with
long-standing type 1 diabetes. Diabetologia 2013.
57. Trebotic LK, Klimek P, Thomas A, Fenzl A, Leitner K, Springer S, Kiefer FW, Kautzky-Willer A:
Circulating Betatrophin Is Strongly Increased in Pregnancy and Gestational Diabetes
Mellitus. PLoS One 2015, 10:e0136701.
58. Li S, Liu D, Li L, Li Y, Li Q, An Z, Sun X, Tian H: Circulating Betatrophin in Patients with Type 2
Diabetes: A Meta-Analysis. J Diabetes Res 2016, 2016:6194750.
This article is protected by copyright. All rights reserved.
59. Erol O, Ellidag HY, Ayik H, Ozel MK, Derbent AU, Yilmaz N: Evaluation of circulating
betatrophin levels in gestational diabetes mellitus. Gynecol Endocrinol 2015, 31:652-656.
60. Wawrusiewicz-Kurylonek N, Telejko B, Kuzmicki M, Sobota A, Lipinska D, Pliszka J,
Raczkowska B, Kuc P, Urban R, Szamatowicz J, et al: Increased Maternal and Cord Blood
Betatrophin in Gestational Diabetes. PLoS One 2015, 10:e0131171.
61. Xie X, Gao H, Wu S, Zhao Y, Du C, Yuan G, Ning Q, McCormick K, Luo X: Increased Cord Blood
Betatrophin Levels in the Offspring of Mothers with Gestational Diabetes. PLoS One 2016,
11:e0155646.
62. Ebert T, Kralisch S, Wurst U, Lossner U, Kratzsch J, Bluher M, Stumvoll M, Toenjes A,
Fasshauer M: Betatrophin levels are increased in women with gestational diabetes mellitus
compared to healthy pregnant controls. Eur J Endocrinol 2015.
63. Crujeiras AB, Zulet MA, Abete I, Amil M, Carreira MC, Martinez JA, Casanueva FF: Interplay
of atherogenic factors, protein intake and betatrophin levels in obese-metabolic syndrome
patients treated with hypocaloric diets. Int J Obes (Lond) 2015.
64. Zhang Y, Li R, Meng Y, Li S, Donelan W, Zhao Y, Qi L, Zhang M, Wang X, Cui T, et al: Irisin
stimulates browning of white adipocytes through mitogen-activated protein kinase p38
MAP kinase and ERK MAP kinase signaling. Diabetes 2014, 63:514-525.
65. Espes D, Lau J, Carlsson PO: Increased levels of irisin in people with long-standing Type 1
diabetes. Diabet Med 2015, 32:1172-1176.
66. Ebert T, Kralisch S, Wurst U, Lossner U, Kratzsch J, Bluher M, Stumvoll M, Tonjes A,
Fasshauer M: Betatrophin levels are increased in women with gestational diabetes mellitus
compared to healthy pregnant controls. Eur J Endocrinol 2015, 173:1-7.
67. Wang L, Song J, Wang C, Lin P, Liang K, Sun Y, He T, Li W, Zhao R, Qin J, et al: Circulating
Levels of Betatrophin and Irisin Are Not Associated with Pancreatic beta-Cell Function in
Previously Diagnosed Type 2 Diabetes Mellitus Patients. J Diabetes Res 2016,
2016:2616539.
68. Fu Z, Abou-Samra AB, Zhang R: A lipasin/Angptl8 monoclonal antibody lowers mouse
serum triglycerides involving increased postprandial activity of the cardiac lipoprotein
lipase. Sci Rep 2015, 5:18502.
69. Calan M, Yilmaz O, Kume T, Unal Kocabas G, Yesil Senses P, Senses YM, Temur M, Gursoy
Calan O: Elevated circulating levels of betatrophin are associated with polycystic ovary
syndrome. Endocrine 2016.
This article is protected by copyright. All rights reserved.
Table 1: A list of studies measuring the level of ANGPTL8 in people with obesity and/or
diabetes
Study
Effect of Obesity
Effect of Diabetes
Comment
Ren et al.[22]
Increased in ob/ob
mice
NA
RIFL gene expression was increased
in fat and liver of obese mice. RIFL
expression was induced by insulin
[22].
Zhang et al.
[25]
Increased
ANGPTL8 in
Obesity.
NA
Expression in mice [25].
Yi et al. [19]
Higher in the liver of
ob/ob mice.
Higher in the liver of
db/db mice.
Gene expression was increased by a
3- to 4-fold in the liver of both ob/ob
and db/db mice. Given the name
betatrophin. Its expression was also
induced by insulin resistance
increasing beta-cell proliferation and
mass [19].
Espes et al.
[56]
NA
Increased
ANGPTL8 level in
people with T1D.
The level of ANGPTL8 from plasma
samples in 33 people with T1D and
24 age-matched healthy controls.
Increased ANGPTL8 in T1D did not
protect against loss of C-peptide [56].
Phoenix Pharmaceuticals ELIS kit.
Abu-Farha et
al [20, 34]
Increased
ANGPTL8 in obese
people.
Increased
ANGPTL8 in people
with T2D.
Used a large cohort of 1049 non-
diabetic people and 556 people with
T2D [20]. Also showed that exercise
reduced ANGGPTL8 level in obese
people [34]. EIAAB ELISA kit.
Fu et al. [36]
Increased
ANGPTL8 level in
obesity.
Increased
ANGPTL8 level in
people with T2D.
15 non-diabetic people vs 14 people
with T2D subjects matched for BMI-
and age. 24 lean vs 29 overweight
and obese people [36]. Phoenix
Pharmaceuticals ELIS kit.
Gomez-
Ambrosi et al.
[39]
Decreased
ANGPTL8 level in
obese people.
Decreased
ANGPTL8 level in
people with T2D.
75 normo-glycemic people, 30 people
with impaired glucose tolerance and
15 people with T2D. Matched for
sex, age and adiposity. The
comparison group comprised 33
normo-glycemic lean people. A
different ELISA kit was used [39].
Cusabio ELISA kit.
Hu et al. [53]
NA
Higher ANGPTL8
level in newly
diagnosed people
with T2D.
83 newly diagnosed people with T2D
compared with83 age, sex and BMI
matched healthy controls [53].
EIAAB ELISA kit.
This article is protected by copyright. All rights reserved.
Yamada et al.
[55]
NA
Higher ANGPTL8
in people with T1D
and people with T2D
compared with
healthy controls.
12 healthy controls, 34 people with
T1D and 30 people with T2D [55].
EIAAB ELISA kit.
Guo et al. [37]
Higher ANGPTL8
level in overweight
people compared
with lean people.
Obese people had
lower ANGPTL8
than overweight
people. Higher
ANGPTL8 level in
overweight people
with T2D. No
significant
difference between
people with T2D
those without
diabetes.
60 people with normal glucose
tolerance: 17 lean, 23 overweight,
and 20 obese. 56 people with T2D:
14 lean, 23 overweight, and 19 obese
[37]. Aviscera Bioscience ELISA kit.
This article is protected by copyright. All rights reserved.
Table 2: A list of studies of the ANGPTL8 level in pregnant women.
Study
Effect of Obesity
Effect of Diabetes
Comment
Ebert et al.
[62].
NA
ANGPTL8 level was
higher in women
with GDM
compared with
healthy pregnant
women. ANGPTL8
level was decreased
postpartum
compared with its
level during
pregnancy.
74 women diagnosed with GDM vs.
74 normo-glycemic pregnant women
matched for gestational age [62].
Phoenix Pharmaceuticals ELIS kit.
Trebotic et al.
[57]
NA
Increased
ANGPTL8 in
pregnant women
with GDM
compared with
pregnant normo-
glycemic women.
21 women with GDM and 19 BMI
matched normo-glycemic pregnant
women [57]. EIAAB ELISA kit.
Erol et al. [59].
NA
Higher ANGPTL8
level in the GDM
women than normo-
glycemic pregnant
women.
45 women with GDM compared
with 45 normo-glycemic pregnant
women matched for BMI and
gestational age [59]. EIAAB ELISA
kit
Wawrusiewicz
-Kurylonek et
al. [60]
NA.
Increased maternal
and cord blood
ANGPTL8 level in
GDM women.
93 women with GDM and 97
normo-glycemic women. Cord
blood ANGPTL8 level and gene
expression was evaluated in 20
women with GDM and 20 normo-
glycemic pregnant women [60].
USCN Life Science ELISA kit.
Xie et al. [61]
NA
Higher ANGPTL8
in cord blood of
women with GDM
than in normo-
glycemic women.
A total of 54 pregnant women who
delivered by Caesarean section: 21
with GDM and 23 normo-glycemic.
ANGPTL8 level was measured in
cord blood [61]. EIAAB ELISA kit.
... It stimulates glycogen synthesis and inhibits gluconeogenesis in cultured hepatocytes, 7 and it also inhibits lipoprotein lipase in muscle, whereas it stimulates it in adipose tissue. 8,9 Visfatin is produced by various tissues in addition to adipose tissue, including liver and skeletal muscle. Its effect on glucose homeostasis is complex because it acts as an extracellular inflammatory mediator, and it also has an important insulin sensitizing effect, 10 possibly by binding to the insulin receptor at a site distinct from insulin. ...
... 11 Obesity results in alterations in adipokine secretion. Betatrophin concentrations are largely increased in obesity in rodents and humans, 8,12 whereas reports regarding visfatin in obesity are inconsistent. 13 Most studies in humans describe increased visfatin concentrations, 11,14 whereas studies in mice show decreased adipose tissue expression. ...
Article
Full-text available
Background Hypothyroidism in dogs is associated with obesity and altered lipid and carbohydrate metabolism. The adipokines, visfatin, and betatrophin, affect glucose tolerance. Betatrophin is involved in lipid regulation. Hypothesis Visfatin and betatrophin serum concentrations are altered in hypothyroid dogs. Animals Dogs with naturally occurring hypothyroidism (n = 25) and healthy dogs (n = 25). Methods Insulin, visfatin, and betatrophin serum concentrations were measured in all dogs and 19 of the hypothyroid dogs after 30 days of thyroxine treatment. Body condition score (BCS) was determined (1‐9 scale). Results Visfatin concentrations were lower in hypothyroid compared with healthy dogs (mean, 95% confidence interval [CI]; 2.0 ng/mL, 1.2‐3.3 vs 5.1 ng/mL, 3.3‐7.8; P = .004) and increased post‐treatment (3.1 ng/mL, 1.9‐4.9 vs 2.6 ng/mL, 1.6‐4.1; P = .05). Betatrophin concentrations were lower in lean to normal (body condition score [BCS], 3‐5) hypothyroid dogs compared to lean to normal healthy dogs (52 pg/mL, 9‐307 vs 597 pg/mL, 216‐1648; P = .03), but were not different between overweight (BCS, 6‐9) hypothyroid and healthy dogs (341 pg/L, 168‐695 vs 178 pg/mL, 77‐415; P = .26), and decreased post‐treatment in overweight dogs (206 pg/mL, 87‐488 vs 268 pg/mL, 112‐640; P = .004). Visfatin concentrations were higher in overweight compared with lean to normal dogs (4.7 ng/mL, 3.3‐6.6 vs 2.2 ng/mL, 1.2‐4.2; P = .04). Betatrophin concentrations were positively correlated with BCS (r = .47, P = .02) and insulin concentrations (r = .48, P = .03) in hypothyroid dogs and negatively correlated with BCS (r = −.47, P = .02) and thyroid stimulating hormone concentrations (r = −.56, P = .01) in healthy dogs. Conclusions and Clinical Importance Hypothyroidism in dogs is associated with alterations in visfatin and betatrophin concentrations that partially resolve with thyroxine treatment.
... Several studies have reported that ANGPTL8 acts as a key modulator in lipid metabolism and metabolic disorders [12,13]. ANGPTL8 has at least two physiological roles, namely those of regulating plasma TG levels and controlling inflammation. ...
... ANGPTL8 plays an important role in lipid and glucose metabolism and is widely associated with various metabolic disorders, such as obesity and T2D, with more recent studies highlighting its role in inflammation [13,36]. However, the role of the ANGPTL8 rs2278426 (p.R59W) variant remains unclear and needs to be explored in the context of inflammation. ...
Article
Full-text available
Background: Angiopoietin-like protein 8 (ANGPTL8) is known to regulate lipid metabolism and inflammation. It interacts with ANGPTL3 and ANGPTL4 to regulate lipoprotein lipase (LPL) activity and with IKK to modulate NF-κB activity. Further, a single nucleotide polymorphism (SNP) leading to the ANGPTL8 R59W variant associates with reduced low-density lipoprotein/high-density lipoprotein (LDL/HDL) and increased fasting blood glucose (FBG) in Hispanic and Arab individuals, respectively. In this study, we investigate the impact of the R59W variant on the inflammatory activity of ANGPTL8. Methods: The ANGPTL8 R59W variant was genotyped in a discovery cohort of 867 Arab individuals from Kuwait. Plasma levels of ANGPTL8 and inflammatory markers were measured and tested for associations with the genotype; the associations were tested for replication in an independent cohort of 278 Arab individuals. Impact of the ANGPTL8 R59W variant on NF-κB activity was examined using approaches including overexpression, luciferase assay, and structural modeling of binding dynamics. Results: The ANGPTL8 R59W variant was associated with increased circulatory levels of tumor necrosis factor alpha (TNFα) and interleukin 7 (IL7). Our in vitro studies using HepG2 cells revealed an increased phosphorylation of key inflammatory proteins of the NF-κB pathway in individuals with the R59W variant as compared to those with the wild type, and TNFα stimulation further elevated it. This finding was substantiated by increased luciferase activity of NF-κB p65 with the R59W variant. Modeled structural and binding variation due to R59W change in ANGPTL8 agreed with the observed increase in NF-κB activity. Conclusion: ANGPTL8 R59W is associated with increased circulatory TNFα, IL7, and NF-κB p65 activity. Weak transient binding of the ANGPTL8 R59W variant explains its regulatory role on the NF-κB pathway and inflammation.
... 2 This increase has been attributed to several factors, including changes in diet with the substitution of animal products and refined foods, 3 and socioeconomic factors, such as an increase in affluence, which reveals a rise in the genetic or ethnic predisposition for diabetes. Mostly diabetes mellitus is linked with a higher risk of cardiovascular complications 4 and doubles the risk of cardiovascular disease (CVD), particularly among individuals with an antiquity of diabetes as opposed to those newly diagnosed with the condition. High BMI also alters hemodynamics and cardiac structure, making it one of the independent risk factors for CVD in T2DM. 5 Independent risk factors for CVD include high cholesterol levels, low-density lipoprotein (LDL-C), and triglycerides (TG) in T2DM. 5 ANGPTL8 is a protein (previously known as betatrophin) that is synthesized in adipose tissues and the liver and discharged into the bloodstream to stimulate cell growth. ...
Article
Full-text available
Purpose This study examines whether Angiopoietin Like 8 (ANGPTL8) is linked to cardiometabolic risk factors (CMRFs) in Saudi women with type 2 diabetes (T2DM). Methods Case-control investigation compared 150 women aged 30–60 with T2DM to 140 healthy women of the same age and gender. Results ANGPTL8 levels differed significantly between T2DM and non-diabetics. Fasting blood glucose (FBG), insulin resistance (IR), triglycerides (TG), high-sensitivity C-reactive protein (hs-CRP), body mass index (BMI), and atherogenic index (AIP) of plasma all correlated positively with ANGPTL8 concentrations. Insulin levels correlated negatively with ANGPTL8. Multiple linear regression models showed that elevated ANGPTL8 independently predicted higher FBG, hs-CRP, IR, TG, and AIP in T2DM patients. Conclusion The study found a significant association between ANGPTL8 levels and IR, hs-CRP, TG, AIP, and BMI in women with T2DM. These components are classified as CMRFs and have the potential to contribute to the development of cardiovascular disease (CVD).
... The study found that a short-duration exercise program reduced betatrophin concentrations in 62 obese persons from 82 non-obese participants who participated in a mix of strength training and moderate-intensity aerobic exercise (Abu-Farha et al., 2016). Long-term physical activity regimens can lower betatrophin concentrations in obese individuals (Abu-Farha et al., 2017). According to a previous study, moderate-intensity exercise had a positive effect on betatrophin levels in obese individuals . ...
Article
Full-text available
Obesity is associated with increased betatrophin levels, which can lead to fat metabolism disorders. Increased levels of betatrophin may inhibit the performance of the lipoprotein lipase (LPL) enzyme, making it difficult for triglycerides (TG) in the blood to be converted into energy and causing excessive fat accumulation in adipose tissue. Physical exercise has been reported to increase energy expenditure in obesity. This study aims to determine changes in betatrophin in obese women after moderate-intensity aerobic exercise. A total of twenty obese women aged 20-30 years took part in the study. The participants were administered into two groups: control (K1) and moderate-intensity aerobic exercise (K2). Moderate-intensity aerobic exercise (60-70% HRmax) was performed five times a week for two weeks. Pretest and posttest betatrophin levels were measured using the Enzyme-Linked Immunosorbent Assay (ELISA) Kit method. Data were analyzed using a paired samples t-test with a significance level of p ≤ 0.05. The results showed the average pretest and posttest betatrophin levels in K1 (0.44±0.14 vs 0.44±0.13 ng/mL; p=0.894), and K2 (0.41±0.05 vs 0.31±0.03 ng/mL; p=0.000). According to the study's findings, moderate-intensity aerobic exercise (60-70% HRmax) performed five times per week for two weeks positively impacted alterations in betatrophin levels in obese women.
... Despite its lack of direct involvement in angiogenesis, ANGPTL8 has been identified as a significant contributor to regulating metabolism processes by adjusting glucose and lipid levels, in addition to its crucial role in maintaining lipid balance [14]. In addition to its association with T2D [16,17], ANGPTL8 is linked to diabetes complications and other concomitant disorders such as DN [15]. Chen et al. reported that ANGPTL8 is considerably higher in T2D patients with various stages of albuminuria [18]. ...
Article
Full-text available
Angiopoietins are crucial growth factors for maintaining a healthy, functional endothelium. Patients with type 2 diabetes (T2D) exhibit significant levels of angiogenic markers, particularly Angiopoietin-2, which compromises endothelial integrity and is connected to symptoms of endothelial injury and failure. This report examines the levels of circulating angiopoietins in people with T2D and diabetic nephropathy (DN) and explores its link with ANGPTL proteins. We quantified circulating ANGPTL3, ANGPTL4, ANGPTL8, Ang1, and Ang2 in the fasting plasma of 117 Kuwaiti participants, of which 50 had T2D and 67 participants had DN. The Ang2 levels increased with DN (4.34 ± 0.32 ng/mL) compared with T2D (3.42 ± 0.29 ng/mL). This increase correlated with clinical parameters including the albumin-to-creatinine ratio (ACR) (r = 0.244, p = 0.047), eGFR (r = −0.282, p = 0.021), and SBP (r = −0.28, p = 0.024). Furthermore, Ang2 correlated positively to both ANGPTL4 (r = 0.541, p < 0.001) and ANGPTL8 (r = 0.41, p = 0.001). Multiple regression analysis presented elevated ANGPTL8 and ACRs as predictors for Ang2’s increase in people with DN. In people with T2D, ANGPTL4 positively predicted an Ang2 increase. The area under the curve (AUC) in receiver operating characteristic (ROC) analysis of the combination of Ang2 and ANGPTL8 was 0.77 with 80.7% specificity. In conclusion, significantly elevated Ang2 in people with DN correlated with clinical markers such as the ACR, eGFR, and SBP, ANGPTL4, and ANGPTL8 levels. Collectively, this study highlights a close association between Ang2 and ANGPTL8 in a population with DN, suggesting them as DN risk predictors.
... [24][25][26] Clinical research has demonstrated that serum ANGPTL8 abnormality is closely related to diabetes, fatty liver, obesity, and metabolic syndrome. 7,[27][28][29] Our preliminary research revealed that abnormal elevation in serum ANGPTL8 levels may be related to IR and PCOS.8 Although our findings indicate that ANGPTL8 functional abnormality may be involved in the occurrence and development of metabolic diseases, including PCOS, the mechanisms underlying it remain unknown. ...
Article
Full-text available
Purpose To study the relationship between the single nucleotide polymorphism (SNP) rs2278426 in the angiopoietin-like protein 8 gene (ANGPTL8) and polycystic ovary syndrome (PCOS). Patients and methods A total of 122 patients with PCOS and 108 controls were recruited for comparison of glucose, lipid, insulin, sex hormone, and ANGPTL8 levels. Polymerase chain reaction (PCR) and gene sequencing were performed for comparison of the frequency of the CC, CT, and TT rs2278426 genotypes and the rs2278426 allele distributions between the PCOS and control groups and between the obese and non-obese subgroups of the PCOS and control groups. Results The frequency of the T allele was significantly higher in the PCOS group than that in the controls (P = 0.037). In the dominant genetic model, the proportion of the CT+TT genotype in the PCOS group was significantly higher than that in the controls (P = 0.047). Subgroup analysis demonstrated that the T allele proportion was significantly higher in obese PCOS group than obese control group (P = 0.027). PCOS with the CT+TT genotype had significantly higher body mass index (BMI; P = 0.001), triglyceride (TG; P = 0.005), homeostasis model assessment of insulin resistance (HOMA-IR; P = 0.035), testosterone (P = 0.041), and ANGPTL8 (P = 0.037) levels and significantly lower high-density lipoprotein (HDL) levels (P = 0.025) than PCOS with the CC genotype. Obese PCOS group with the CT+TT genotype had significantly higher TG (P = 0.015), luteinizing hormone (LH; P = 0.030), fasting insulin (FINS; P = 0.039), HOMA-IR (P = 0.018), and ANGPTL8 (P = 0.049) levels than obese PCOS group with the CC genotype. Conclusion Polymorphisms of rs2278426 may induce glycolipid metabolic disorders by affecting ANGPTL8 levels and functions in Han Chinese females with obesity from the Shandong region, increasing the risk of PCOS in this population.
... Betatrophin is produced in the liver and adipocytes that are two crucial tissues involved in insulin signaling pathways. The abnormal buildup of fat tissue not only leads to insulin resistance but also disrupts the production of betatrophin 8 . ...
Article
Full-text available
Central adiposity presents an important risk factor for advancing insulin insensitivity and type 2 diabetes mellitus. Betatrophin, a liver or adipocyte-derived hormone, was assumed to improve islet insulin secretion and compensate insulin resistance but its level during obesity is still conflicted. This study aimed to explore serum betatrophin level in centrally-obese middle-aged men with diabetic potentials compared with age-matched non-obese ones. Sixty-eight male subjects of 40-60 years of age, residing in North Okkalapa Township, Yangon, Myanmar, were recruited and classified into centrally-obese group (n=34) and non-obese group (n=34). Fasting blood samples were obtained to quantify plasma glucose by glucose oxidase method, and serum insulin and betatrophin levels by ELISA. Plasma glucose levels were comparable between the two groups, while insulin concentration of obese group was significantly greater than that of non-obese group. Therefore, HOMA-IR was markedly increased in obese subjects when compared to non-obese ones (4.87±0.28 vs 1.90±0.14, p<0.001) and so did HOMA-β (310.88±26.58 vs 149.00±11.83, p<0.001). Interestingly, betatrophin hormone level was significantly reduced in obese group than non-obese group (1.72±0.21 vs 2.72±0.26 ng/ml, p<0.01). Moreover, betatrophin had a strong negative correlation with glucose and insulin levels (p<0.05) as well as with the indicator of central adiposity, waist circumference (p<0.05), among the subjects. However, significant correlation between betatrophin and HOMA-IR and HOMA-β was not observed in both groups (p=0.14 and 0.20 respectively). Taken together, betatrophin hormone has been found to decrease in adult central obesity, which is noticeably associated with insulin resistance and compensatory beta-cell hyperfunction. Betatrophin, previously regarded as beta-cell mitogen, has been denied in this study, owing to lack of correlation with HOMA indexes of diabetes.
... Diabetes mellitus (DM) is characterized by hyperinsulinemia and hyperglycemia (98,99). ANGPTL8, which has a unique characteristic in regulating lipid and glucose metabolism, is upregulated in diabetes and is becoming increasingly recognized as a potential drug target for the treatment of diabetes and related metabolic disorders (100,101). A study showed that the FLOT1 gene was co-expressed with ANGPTL8. ...
Article
Full-text available
FLOT1, a scaffold protein of lipid rafts, is involved in several biological processes, including lipid raft protein-dependent or clathrin-independent endocytosis, and the formation of hippocampal synapses, amongst others. Increasing evidence has shown that FLOT1 can function as both a cancer promoter and cancer suppressor dependent on the type of cancer. FLOT1 can affect the occurrence and development of several types of cancer by affecting epithelial-mesenchymal transition, proliferation of cancer cells, and relevant signaling pathways, and is regulated by long intergenic non-coding RNAs or microRNAs. In the nervous system, overexpression or abnormally low expression of FLOT1 may lead to the occurrence of neurological diseases, such as Alzheimer's disease, Parkinson's disease, major depressive disorder and other diseases. Additionally, it is also associated with dilated cardiomyopathy, pathogenic microbial infection, diabetes-related diseases, and gynecological diseases, amongst other diseases. In the present review, the structure and localization of FLOT1, as well as the physiological processes it is involved in are reviewed, and then the upstream and downstream regulation of FLOT1 in human disease, particularly in different types of cancer and neurological diseases are discussed, with a focus on potentially targeting FLOT1 for the clinical treatment of several diseases.
Article
Full-text available
Aim: Exposing a fetus to hyperglycemia can increase the risk for later-life metabolic disorders. Betatrophin has been proposed as a key regulator of pancreatic beta cell proliferation and lipid regulation. Highly responsive to nutritional signals, serum betatrophin concentrations have been found to be altered by various physiological and pathological conditions. We hypothesized that betatrophin levels are increased in the cord blood in offspring exposed to intrauterine hyperglycemia. Methods: This was a cross-sectional study including 54 mothers who underwent uncomplicated Cesarean delivery in a university hospital. Maternal gestational glucose concentration was determined at 24-48 weeks gestation after a 75-g OGTT. Cord blood and placental tissue was collected immediately post delivery. Metabolic parameters were determined in the Clinical Laboratory. Cord blood betatrophin levels were assayed using a commercially available ELISA kit. Placental mitochondrial content was determined by real-time PCR. Results: Cord blood betatrophin levels were increased in the gestational diabetes mellitus (GDM) group compared with the normoglycemic group. Furthermore, betatrophin levels were positively correlated with maternal gestational 2h post-OGTT glucose, cord blood insulin, HOMA-IR, and inversely correlated with placental mitochondrial content. Conclusions: Cord blood betatrophin may function as a potential biomarker of maternal intrauterine hyperglycemia and fetal insulin resistance, which may presage for long-term metabolic impact of GDM on offspring.
Article
Full-text available
Thyroid hormone (TH) affects many metabolic processes such as promoting oxidation of sugar, fat, and protein in many tissues. Thyroid dysfunction is associated with metabolic disorders. The newly discovered adipocyte- and hepatocyte-derived cytokine, betatrophin, has been reported to be involved in metabolic diseases, but its influence on thyroid dysfunction is uncertain. Therefore, the present study aims to evaluate circulating betatrophin levels in subjects with different thyroid function status and to predict the factors associated with betatrophin levels, especially whether thyroid stimulating hormone (TSH), TH, or thyroid autoantibodies are associated with betatrophin levels. In the study, serum betatrophin was measured in the subjects grouped as overt hypothyroidism (OH), subclinical hypothyroidism (SCH), euthyroid with isolated thyroid peroxidase antibody positivity (isolated Ab), and healthy control (HC), according to their thyroid functions. From our results, we found that betatrophin may be associated with thyroid insufficiency but not thyroid autoimmunity. Thus, when interpreting the results of betatrophin, thyroid functions should also be taken into consideration.
Article
Full-text available
ANGPTL8 (betatrophin) has been recently identified as a regulator of lipid metabolism through its interaction with ANGPTL3. A sequence variant in ANGPTL8 has been shown to associate with lower level of Low Density Lipoprotein (LDL) and High Density Lipoprotein (HDL). The objective of this study is to identify sequence variants in ANGPTL8 gene in Arabs and investigate their association with ANGPTL8 plasma level and clinical parameters. A cross sectional study was designed to examine the level of ANGPTL8 in 283 non-diabetic Arabs, and to identify its sequence variants using Sanger sequencing and their association with various clinical parameters. Using Sanger sequencing, we sequenced the full ANGPTL8 gene in 283 Arabs identifying two single nucleotide polymorphisms (SNPs) Rs.892066 and Rs.2278426 in the coding region. Our data shows for the first time that Arabs with the heterozygote form of (c.194C > T Rs.2278426) had higher level of Fasting Blood Glucose (FBG) compared to the CC homozygotes. LDL and HDL level in these subjects did not show significant difference between the two subgroups. Circulation level of ANGPTL8 did not vary between the two forms. No significant changes were observed between the various forms of Rs.892066 variant and FBG, LDL or HDL. Our data shows for the first time that heterozygote form of ANGPTL8 Rs.2278426 variant was associated with higher FBG level in Arabs highlighting the importance of these variants in controlling the function of betatrophin.
Article
Full-text available
ANGPTL8 also called betatrophin is a regulator of lipid metabolism through its interaction with ANGPTL3. It has also been suggested to play a role in insulin resistance and beta-cell proliferation. Based on its function, we hypothesized that ANGPTL8 will play a role in Metabolic Syndrome (MetS). To test this hypothesis we designed this study to measure ANGPTL8 level in subjects with MetS as well as its association with high sensitivity C-reactive protein (HsCRP) level in humans. ANGPTL8 level was measured using ELISA in subjects with MetS as well as their controls, a total of 1735 subjects were enrolled. HsCRP was also measured and its association with ANGPTL8 was examined. ANGPTL8 level was higher in subjects with MetS 1140.6 (171.9–11736.1) pg/mL compared to 710.5 (59.5–11597.2) pg/mL in the controls. Higher levels of ANGPTL8 were also observed with the sequential increase in the number of MetS components (p value = <0.0001). ANGPTL8 showed strong positive correlation with HsCRP (r = 0.15, p value = <0.0001). Stratifying the population into tertiles according to the level of HsCRP showed increased ANGPTL8 level at higher tertiles of HsCRP in the overall population (p value = <0.0001).A similar trend was also observed in MetS and non-MetS subjects as well as in non-obese and obese subjects. Finally, multiple logistic regression models adjusted for age, gender, ethnicity and HsCRP level showed that subjects in the highest tertiles of ANGPTL8 had higher odds of having MetS (odd ratio [OR] = 2.3, 95 % confidence interval [CI] = (1.6–3.1), p value <0.0001. In this study we showed that ANGPTL8 is increased in subjects with MetS and it was significantly associated with HsCRP levels in different subgroups highlighting its potential role in metabolic and inflammatory pathways.
Article
Full-text available
Betatrophin is a newly identified hormone determined to be a potent inducer of pancreatic beta cell proliferation in response to insulin resistance in mice. Polycystic ovary syndrome (PCOS) is an inflammatory-based metabolic disease associated with insulin resistance. However, no evidence is available indicating whether betatrophin is involved in women with PCOS. The objective of this study was to ascertain whether betatrophin levels are altered in women with PCOS. This study was conducted in secondary referral center. This cross-sectional study included 164 women with PCOS and 164 age- and BMI-matched female controls. Circulating betatrophin levels were measured using ELISA. Metabolic and hormonal parameters were also determined. Circulating betatrophin levels were significantly elevated in women with PCOS compared with controls (367.09 ± 55.78 vs. 295.65 ± 48.97 pg/ml, P < 0.001). Betatrophin levels were positively correlated with insulin resistance marker homeostasis model assessment of insulin resistance (HOMA-IR), free-testosterone, high-sensitivity C-reactive protein (hs-CRP), atherogenic lipid profiles, and BMI in PCOS. Multivariate logistic regression analyses revealed that the odds ratio for PCOS was 2.51 for patients in the highest quartile of betatrophin compared with those in the lowest quartile (95 % CI 1.31–4.81, P = 0.006). Multivariate regression analyses showed that HOMA-IR, hs-CRP, and free-testosterone were independent factors influencing serum betatrophin levels. Betatrophin levels were increased in women with PCOS and were associated with insulin resistance, hs-CRP, and free-testosterone in these patients. Elevated betatrophin levels were found to increase the odds of having PCOS. Further research is needed to elucidate the physiologic and pathologic significance of our findings.
Article
Full-text available
Objective: ANGPTL8 is a liver and adipose tissue produced protein that regulates the level of triglyceride in plasma as well as glucose homeostasis. This study was designed to evaluate the level of ANGPTL8 in obese and non-obese subjects before and after exercise training. Methods: A total of 82 non-obese and 62 adult obese were enrolled in this study. Subjects underwent a three months of exercise training. Both full length and C-terminal 139-198 form of ANGPTL8 were measured by ELISA. Results: Our data show that the full length ANGPTL8 level was increased in obese subjects (1150.04 ± 108.10 pg/mL) compared to non-obese (775.54 ± 46.12) pg/mL (p-Value = 0.002). C-terminal 139-198 form of ANGPTL8 was also increased in obese subjects 0.28 ± 0.04 ng/mL vs 0.20 ± 0.02 ng/mL in non-obese (p-value = 0.058). In obese subjects, the levels of both forms were reduced after three months of exercise training; full length was reduced from 1150.04 ± 108.10 pg/mL to 852.04 ± 51.95 pg/mL (p-Values 0.015) and c-terminal form was reduced from 0.28 ± 0.04 ng/mL to 0.19 ± 0.03 ng/mL (p-Value = 0.058). Interestingly, full length ANGPTL8 was positively associated with fasting blood glucose (FBG) in non-obese (r = 0.317, p-Value = 0.006) and obese subjects (r = 0.346, p-Value = 0.006) C-terminal 139-198 form of ANGPTL8 on the other hand, did not show any correlation in both groups. Conclusion: In conclusion, our data demonstrate that ANGPTL8 was increased in obesity and reduced after exercise training supporting the potential therapeutic benefit of reducing ANGPTL8. The various forms of ANGPTL8 associated differently with FBG suggesting that they have different roles in glucose homeostasis.
Article
Full-text available
Lipasin/Angptl8 is a feeding-induced hepatokine that regulates triglyceride (TAG) metabolism; its therapeutical potential, mechanism of action, and relation to the lipoprotein lipase (LPL), however, remain elusive. We generated five monoclonal lipasin antibodies, among which one lowered the serum TAG level when injected into mice, and the epitope was determined to be EIQVEE. Lipasin-deficient mice exhibited elevated postprandial activity of LPL in the heart and skeletal muscle, but not in white adipose tissue (WAT), suggesting that lipasin suppresses the activity of LPL specifically in cardiac and skeletal muscles. Consistently, mice injected with the effective antibody or with lipasin deficiency had increased postprandial cardiac LPL activity and lower TAG levels only in the fed state. These results suggest that lipasin acts, at least in part, in an endocrine manner. We propose the following model: feeding induces lipasin, activating the lipasin-Angptl3 pathway, which inhibits LPL in cardiac and skeletal muscles to direct circulating TAG to WAT for storage; conversely, fasting induces Angptl4, which inhibits LPL in WAT to direct circulating TAG to cardiac and skeletal muscles for oxidation. This model suggests a general mechanism by which TAG trafficking is coordinated by lipasin, Angptl3 and Angptl4 at different nutritional statuses.
Article
Full-text available
Objective: To investigate the association between circulating betatrophin level and type 2 diabetes mellitus (T2DM) in human. Methods: A comprehensive literature search was performed in PubMed and Embase databases to identify eligible studies assessing the circulating levels of betatrophin in both T2DM patients and nondiabetic adults. Results: A total of nine eligible studies with twelve comparisons were included for the final meta-analysis. Circulating betatrophin levels in T2DM patients were higher than those in the nondiabetic controls (random-effect SMD 0.53; 95% CI 0.13 to 0.94; P = 0.010). In the subgroup of nonobese population but not the obese population, the overall betatrophin level in T2DM patients was much higher than that in the nondiabetic controls (nonobese: random-effect SMD, 0.82; 95% CI 0.42 to 1.21; P < 0.001; obese: random-effect SMD, -0.39; 95% CI, -0.95 to 0.18; P = 0.18). Metaregression indicated that body mass index of T2DM patients was associated with mean difference of betatrophin level between T2DM and nondiabetic adults (slope, -578.8; t = -2.7; P = 0.02). Conclusion: Based on the findings of our meta-analysis, circulating betatrophin level of T2DM patients is higher than that of nondiabetic adults in the nonobese population, but not in the obese population.
Article
Full-text available
Betatrophin and irisin are two recently identified hormones which may participate in regulating pancreatic β -cell function. However, the associations of these two hormones with β -cell function remain unclear. The present study aims to demonstrate the associations of circulating betatrophin and irisin levels with β -cell function, assessed by the area under the curve (AUC) of C-peptide, and the possible correlation between these two hormones in previously diagnosed type 2 diabetes mellitus (T2DM) patients. In total, 20 age-, sex-, and body mass index- (BMI-) matched normal glucose tolerance (NGT) subjects and 120 previously diagnosed T2DM patients were included in this study. Partial correlation analysis was used to evaluate the relationships between these two hormones and indexes of β -cell function and insulin resistance. Our results showed that betatrophin levels were significantly elevated, while irisin levels were significantly decreased, in patients with T2DM compared with NGT subjects. However, partial correlation analysis showed that betatrophin levels did not correlate with β -cell function-related variables or insulin resistance-related variables before or after controlling multiple covariates, while irisin correlated positively with insulin sensitivity but is not associated with β -cell function-related variables. Besides, no correlation was observed between betatrophin and irisin levels. Hence we concluded that betatrophin and irisin were not associated with β -cell function in previously diagnosed T2DM patients.
Article
To evaluate the status of serum betatrophin levels and potential relations between metabolic parameters and betatrophin levels in patients with polycystic ovary syndrome. We included patients newly diagnosed with PCOS in our study. Fifty-seven female patients (30 patients with PCOS and 27 healthy control subjects) were enrolled in this study. Serum betatrophin levels were measured using a betatrophin enzyme-linked immunosorbent assay kit. Insulin resistance was calculated using the homeostasis model of the assessment-insulin resistance index formula. The betatrophin level was 1538,85 ng/L in the patient group and 2440,46 ng/L in the control group, and the difference was statistically significant (p=0.003). A significantly negative correlation was found between betatrophin level and insulin, HOMA-IR, and BMI. Betatrophin levels in patients with PCOS are lower than those without PCOS and inversely related to insulin resistance.