ArticlePDF AvailableLiterature Review

Abstract

Purpose of the review: Diabetes mellitus is a major cause of kidney disease [chronic kidney disease (CKD) and end-stage renal disease (ESRD)] and are both characterized by an increased risk of cardiovascular events. Diabetes and kidney disease are also commonly associated with a chronic inflammatory state, which is now considered a non-traditional risk factor for atherosclerosis. In the case of type 2 diabetes mellitus (T2DM), inflammation is mainly a consequence of visceral obesity, while in the case of CKD or ESRD patients on dialysis, inflammation is caused by multiple factors, classically grouped as dialysis-related and non-dialysis-related. More recently, a key role has been credited to the intestinal microbiota in the pathogenesis of chronic inflammation present in both disease states. While many recent data on the intestinal microbiota and its relationship to chronic inflammation are available for CKD patients, very little is known regarding T2DM and patients with diabetic nephropathy. The aim of this review is to summarize and discuss the main pathophysiological issues of intestinal microbiota in patients with T2DM and CKD/ESRD. Recent findings: The presence of intestinal dysbiosis, along with increased intestinal permeability and high circulating levels of lipopolysaccharides, a condition known as "endotoxemia," characterize T2DM, CKD, and ESRD on dialysis. The hallmark of intestinal dysbiosis is a reduction of saccharolytic microbes mainly producing short-chain fatty acids (SCFA) and, in the case of CKD/ESRD, an increase in proteolytic microbes that produce different substances possibly related to uremic toxicity. Dysbiosis is associated with endotoxemia and chronic inflammation, with disruption of the intestinal barrier and depletion of beneficial bacteria producing SCFAs. T2DM and CKD/ESRD, whose coexistence is increasingly found in clinical practice, share similar negative effects on both intestinal microbiota and function. More studies are needed to characterize specific alterations of the intestinal microbiota in diabetic nephropathy and to assess possible effects of probiotic and prebiotic treatments in this setting.
MICROVASCULAR COMPLICATIONSNEPHROPATHY (M AFKARIAN, SECTION EDITOR)
Intestinal Microbiota in Type 2 Diabetes and Chronic Kidney
Disease
Alice Sabatino
1
&Giuseppe Regolisti
1
&Carmela Cosola
2
&Loreto Gesualdo
2
&
Enrico Fiaccadori
1
#Springer Science+Business Media New York 2017
Abstract
Purpose of the Review Diabetes mellitus is a major cause
of kidney disease [chronic kidney disease (CKD) and end-
stage renal disease (ESRD)] and are both characterized by
an increased risk of cardiovascular events. Diabetes and
kidney disease are also commonly associated with a
chronic inflammatory state, which is now considered a
non-traditional risk factor for atherosclerosis. In the case
of type 2 diabetes mellitus (T2DM), inflammation is
mainly a consequence of visceral obesity, while in the
case of CKD or ESRD patients on dialysis, inflammation
is caused by multiple factors, classically grouped as
dialysis-related and non-dialysis-related. More recently, a
key role has been credited to the intestinal microbiota in
the pathogenesis of chronic inflammation present in both
disease states. While many recent data on the intestinal
microbiota and its relationship to chronic inflammation
are available for CKD patients, very little is known re-
garding T2DM and patients with diabetic nephropathy.
The aim of this review is to summarize and discuss the
main pathophysiological issues of intestinal microbiota in
patients with T2DM and CKD/ESRD.
Recent Findings The presence of intestinal dysbiosis, along
with increased intestinal permeability and high circulating
levels of lipopolysaccharides, a condition known as
endotoxemia,characterize T2DM, CKD, and ESRD on di-
alysis. The hallmark of intestinal dysbiosis is a reduction of
saccharolytic microbes mainly producing short-chain fatty
acids (SCFA) and, in the case of CKD/ESRD, an increase in
proteolytic microbes that produce different substances possi-
bly related to uremic toxicity.
Summary Dysbiosis is associated with endotoxemia and
chronic inflammation, with disruption of the intestinal barrier
and depletion of beneficial bacteria producing SCFAs. T2DM
and CKD/ESRD, whose coexistence is increasingly found in
clinical practice, share similar negative effects on both intes-
tinal microbiota and function. More studies are needed to
characterize specific alterations of the intestinal microbiota
in diabetic nephropathy and to assess possible effects of pro-
biotic and prebiotic treatments in this setting.
Keywords Chronic kidney disease .Diabetes mellitus .
Endotoxin .Inflammation .Intestinal microbiota .Uremia
Introduction
Type 2 diabetes mellitus (T2DM) accounts for at least 90% of all
diabetes cases in the adult population [1]. In the last two decades,
a true epidemic of T2DM has been observed, with more than
300 million people being affected globally [2]. It is estimated that
more than 80% of patients with T2DM are overweight or have
obesity, which is now considered the main cause of the disease
[3]. Diabetes mellitus is also the leading cause of chronic kidney
disease (CKD) and end-stage renal disease (ESRD), non-
traumatic limb amputation, and blindness among adults [1]. In
This article is part of the Topical Collection on Microvascular
ComplicationsNephropathy
*Enrico Fiaccadori
enrico.fiaccadori@unipr.it
1
Unità di Fisiopatologia dellInsufficienza Renale Acuta e Cronica,
Università degli Studi di Parma, Parma, Italy
2
Dipartimento dellEmergenza e dei Trapianti di OrganiSezione di
Nefrologia, Dialisi e Trapianti, Università degli Studi di Bari Aldo
Moro, Bari, Italy
Curr Diab Rep (2017) 17:16
DOI 10.1007/s11892-017-0841-z
the USA, T2DM is the primary cause of ESRD, being responsi-
ble for 44% of all new cases in 2011 [4].
CKD is a global health issue since 610% of the whole adult
population can be diagnosed with the disease according to the
most recent classification [5,6]. In this clinical setting, the most
frequent cause of death is cardiovascular disease (CVD), which
is attributable to the coexistence of traditional (e.g., hypertension,
diabetes, and dyslipidemia) and non-conventional risk factors
[7]. Among the latter, persistent low-grade inflammation has
received increasing attention, and it is now regarded as a major
catalyst for CVD in CKD [7]. Chronic inflammation is usually
defined as a persistent inflammatory response by a causative
stimulus. In the case of CKD, and especially ESRD on dialysis,
increased production and decreased renal clearance contribute to
the accumulation of cytokines [7]. The evidence concerning the
use of inflammatory markers to diagnose chronic inflammation
in the course of ESRD is vast [8] and suggests interleukin 6 (IL-
6) as an important marker of inflammation, also representing the
best outcome predictor in advanced CKD and ESRD [9].
However, since IL-6 measurement is not easily available in the
clinical practice, the assessment of C-reactive protein (CRP), a
marker of systemic inflammation and predictor of cardiovascular
risk, is now the standard of care in the clinical setting because of
its reliability, low cost, and wide availability [10].
Chronic inflammation is common in patients with T2DM,
CKD, and ESRD [7,1119]. In the case of T2DM, inflamma-
tion is mainly considered a consequence of obesity, in partic-
ular visceral obesity [2,20,21]. As for CKD and ESRD, many
dialysis-related and non-dialysis-related factors are thought to
contribute to the chronic inflammatory state [10,22], and re-
cent research has also highlighted a key role of the intestinal
microbiota. Two main pathophysiological mechanisms are
likely to be involved. Firstly, the low-grade inflammation typ-
ical of T2DM and CKD and ESRD can be potentiated by
translocation, from the gut lumen to the blood, of bacteria
and bacterial products (e.g., lipopolysaccharides, LPS) caused
by an increase in intestinal permeability (leaky gut syn-
drome)[12,13,2327]. Secondly, modifications in the intes-
tinal microbiota in terms of species richness, diversity, com-
position, and function may have a profound impact on host
physiology, through changes in nutrient utilization and syn-
thesis of bioactive metabolites [11,27]. While abundant evi-
dence has been accrued recently in patients with CKD or
ESRD, information on this issue in T2DM is scarce. Thus,
this review is aimed at summarizing and discussing the main
pathophysiology of the intestinal microbiota in the presence of
T2DM and CKD and ESRD.
Intestinal Microbiota in Healthy Subjects
In humans, microbial cells from different bacterial species out-
number human cells by 10-fold, with the gastrointestinal (GI)
tract being the habitat for greater than 70% of this microbial
population [28]. The amount of microbes changes along the
intestine, being highest in its distal tract, where the environment
is poor in oxygen and rich in molecules that these micro-
organisms can utilize as nutrients [28]. The intestinal microbiota
exerts important trophic and protective functions that are not
limited to the intestine but can affect the whole organism
(Table 1). The composition, function, and structure of the intes-
tinal microbiota is generally stable, but it is also very adaptive
depending on the biochemical environment of the GI tract and
changes in nutrient availability, which represents the most impor-
tant regulator of bacterial metabolism [28]. Normal or healthy
intestinal microbiota consists of the bacterial phyla Firmicutes
and Bacteroidetes (>90%), followed by Actinobacteria and
Verrucomicrobia; usually few (0.1%) pathogenic and opportu-
nistic species are present [2931].
The two main nutrients utilized by intestinal bacteria are
carbohydrates and proteins. The ratio between these two mac-
ronutrients significantly impacts on the predominance of dif-
ferent species. In the presence of adequate amounts of undi-
gested complex carbohydrates (especially dietary fibers),
saccharolytic bacteria are favored, and proteins are used for
bacterial growth, while carbohydrates are used for energy pro-
duction through bacterial anaerobic metabolism (fermenta-
tion). As a consequence of carbohydrate fermentation, meth-
ane, hydrogen, and short-chain fatty acids (SCFA) are pro-
duced as end-products [27]. However, in the absence of undi-
gested carbohydrates, also proteins and other nitrogen
Tabl e 1 Physiologic effects of gut microbiota
(A)Integrity and function of GI tract
Maintenance of tight junction protein structure
Induction of epithelial heat-shock proteins
Upregulation of mucin genes
Competition with pathogenic bacteria for binding to intestinal epithelial
cells
Secretion of antimicrobial peptides
Suppression of intestinal inflammation
(B)Immunologic effects
Maturation of intestinal immune system
Reduction of allergic response to food and environmental antigens
Promotion of immunomodulation and cell differentiation
(C)Metabolic effects
Breakdown of indigestible plant polysaccharides and resistant starch
Facilitated absorption of complex carbohydrates
Synthesis of vitamins (K and B groups)
Synthesis of amino acids (threonine and lysine)
Biotransformation of conjugated bile acids
Degradation of dietary oxalates
From: Sabatino A et al. Nephrol Dial Transplant, 2015, 30:924933, by
permission of Oxford University Press [32]
GI gastrointestinal
16 Page 2 of 9 Curr Diab Rep (2017) 17:16
compounds can be fermented by proteolytic bacteria in order
to increase energy availability, with parallel production of po-
tentially toxic end-products, such as ammonia, amines, thiols,
phenols, and indoles [27].
Derangements of the intestinal milieu and related changes
in the composition of the intestinal microbiota represent a
condition referred to as intestinal dysbiosisthat may trigger
a systemic inflammatory response [28,32]. Many external
(e.g., antibiotics and nutrient intake) and internal (e.g., host
genotype, extra-intestinal non-communicable diseases, and
inflammatory bowel diseases) factors may contribute to the
pathogenesis of intestinal dysbiosis and to the overgrowth of
pathobionts (microbes with pathogenic potential) [32].
Normally, the intestinal barrier prevents the translocation of
substances and microbes from the lumen to the bloodstream. The
intestinal barrier is formed by different structures/systems: tight
junctions, enterocyte membranes, mucus secretion, and immu-
nologic defense mechanisms in the intestinal wall [28,33••].
Particularly, tight junctions are a very efficient mechanical pro-
tection against the translocation of substances and bacteria along
para-cellular pathways from the gut to the bloodstream; in fact,
they bind together with epithelial cells and are capable of
adjusting their tightness according to physiological needs [34].
Intestinal Microbiota in Type 2 Diabetes
Obesity-induced insulin resistance is the dominant underlying
pathophysiological factor for the development of T2DM [3].
Obesity is a state of chronic low-grade systemic inflammation,
which is a well-known cause of insulin resistance [35].
In mouse models of obesity, dysbiosis is usually present
[17,36,37]. Specifically, a decrease in the Bacteroidetes/
Firmicutes ratio is associated with the obese state. Germ-free
mice are resistant to obesity induced by a high-fat diet [36],
and colonization of germ-free mice with the microbiota of
obese female humans caused obesity in the colonized mouse
[38], indicating that the composition of the microbiota can
predispose to the development of obesity. In addition, cohous-
ing the colonized mice with lean mice and giving them a low-
fat and high-fiber diet prevented further increase in adiposity,
while in lean animals no changes toward obesity were found,
suggesting that in the end, diet is responsible for phenotype
development [39].
A recent human study demonstrated that poor diversity of
intestinal microbiota (defined as low gene count, LGC) is also
associated with obesity, insulin resistance, hepatic steatosis,
and low-grade inflammation [11]. In this study, LGC subjects
had a more pro-inflammatory microbial profile, characterized
by a reduction of butyrate-producing bacteria and increased
mucus degradation and oxidative stress [11].
Similar results were found in the two largest metagenome
studies in T2DM [13,40]. A moderate dysbiosis was
demonstrated, characterized by a microbiota with decreased
butyrate synthesis capacity [13,40]. Dysbiosis promoted en-
richment in membrane transport of sugars and branched chain
amino acids, and increase in oxidative stress response and
sulfate reduction [13]. Table 2summarizes the major findings
from studies in T2DM patients. Earlier studies in humans and
in mice models of T2DM and obesity reported that obesity
and impaired glucose metabolism were associated with an
altered microbiota in comparison to healthy subjects [12,41,
42]. Particularly, a proliferation of Gram-negative bacteria
might explain the increase in serum LPS levels in obese and
T2DM patients, likely triggering the low-grade inflammation
state typical of these two conditions [12,13].
Indeed, endotoxemia is known to induce the secretion of
pro-inflammatory cytokines [14]. Studies on animal models
and humans have demonstrated that a high-fat diet is able to
modulate intestinal microbiota and to increase serum levels of
LPS. The mechanisms involved in this endotoxemia state are
probably related to an increased uptake of LPS in chylomi-
crons secreted from enterocytes and an increased intestinal
permeability, known as leaky gut[1417]. Circulating
LPS are recognized by Toll-like receptors and activate the
innate immune system and pro-inflammatory pathways.
Glucose and energy metabolism are also influenced by the
microbial production of SCFA. Butyrate is the main source of
energy to the intestinal epithelium and also seems to have an
effect on insulin sensitivity and energy balance [43], while ace-
tate and propionate are mainly substrates for gluconeogenesis
and lipogenesis in the liver. In addition, butyrate has been dem-
onstrated to increase the secretion of GLP1 and PYY from L
cells in the colon [43,44] and to increase the intestinal transit
time [45]. Furthermore, GLP-1 and the activation of the complex
GLP-1/GLP-1 receptor have been demonstrated to ameliorate
the early effects of diabetes on the kidney in part by attenuating
proximal tubular hyper-reabsorption and growth [46].
To summarize, present evidence suggests that dysbiosis
may result in a leaky gut syndrome,with increased perme-
ability that might activate the innate immune system, altering
signaling pathways that affect lipid and glucose metabolism
and triggering low-grade inflammation, eventually leading to
insulin resistance and possibly T2DM. However, studies in
Tabl e 2 Major findings from studies in patients with T2DM
Reduced butyrate-producing bacteria (Roseburia intestinalis and
Faecalibacterium prausnitzii)
Moderate dysbiosis
Pro-inflammatory environment with increased expression of microbial
genes involved in oxidative stress
Reduced expression of genes involved in vitamin synthesis
Increased serum LPS concentration
Increased intestinal permeability
LPS lipopolysaccharides, T2DM type 2 diabetes mellitus
Curr Diab Rep (2017) 17:16 Page 3 of 9 16
human are yet to fully ascertain whether dysbiosis is a cause or
consequence of T2DM [47].
Intestinal Microbiota in CKD and ESRD
The causeconsequence relationship between the alterations of
intestinal microbiota and kidney disease is complex and diffi-
cult to dissect. Indeed, it is reasonable to hypothesize that both
factors may influence each other. A dysbiotic microbiota seems
to represent a susceptibility factor for the development of kid-
ney disease following injury or in predisposed individuals [48].
In addition, it is known that the progressive loss of kidney
function significantly contributes to worsen the intestinal
dysbiosis found in CKD/ESRD patients [49]. Different mech-
anisms are involved due to derangement of the intestinal barrier
and modifications of microbiota composition (Table 3).
In this context, specific associations have been recently
shown between certain intestinal [50] and salivary [51]micro-
bial phyla/families and the condition of IgA nephropathy in
comparison to healthy controls, with a further discrimination
between progressor and non-progressor patients. This evi-
dence suggests an active involvement of microbiota in the
etiology and/or progression of this particular kidney disease,
characterized by the key involvement of the immune system,
which is known to be importantly influenced and modulated
by the microbiota [21]. In this complicated framework, one
additional variable to be considered is the diet, known to sub-
stantially contribute to the modifications of intestinal microbi-
ota composition and metabolism [52]. It is by now ascertained
that typical dietary restrictions, used to be considered as man-
datory for the conservative treatment of CKD, are responsible
of the worsening of the intestinal dysbiosis already occurring
in this clinical setting. Dietary fiber is the primary substrate for
colonic bacterial fermentation; however, patients with CKD
often have a low intake ofdietary fibers, mainly because of the
need to control potassium intake. The main sources of potas-
sium and fiber in the diet are fruits and vegetables, which are
reportedly low in CKD patientsdiet [53]. Reduced intake of
fibers, in addition to other factors related to CKD treatment
(dialysis modality, phosphate binders, low fluid intake), life-
style (inactivity), and comorbidities (diabetes, malnutrition,
heart failure, and cerebrovascular diseases), may prolong the
GI transit time. Prolonged transit time may lead to increased
CHO fermentation in the proximal segments of the intestine
[54], thus reducing CHO availability to the colonic bacteria. In
addition, protein digestion and absorption seem tobe impaired
in CKD patients [55] due to alterations in the GI tract motility,
hypochlorhydria, and bacterial overgrowth in the small bowel,
thus increasing the amount of intact protein available for pro-
teolytic bacteria in the colon [54,56,57].
Recent concepts about the nutritional management of CKD
are expanding the vision from the focus on a single nutrient to
the concept of food matrices,namely complex associations
of different food categories, often nutraceuticals (antioxidants,
fibers, proteins, vitamins, etc.) found together in the same
food. In particular, food matrices traditionally belonging to
the Mediterranean Diet are particularly rich in nutraceutical
components so that this dietary scheme is being reconsidered
as suitable for this category of patients [58].
As discussed earlier, the intestinal microbiota can be highly
adaptable to changes in the biochemical environment. That is
why relevant quantitative and qualitative changes in the bac-
terial population of CKD patients have been demonstrated,
also in earlier stages of the syndrome [59]. Recent studies
have demonstrated increased counts of aerobic and anaerobic
bacteria in the small bowel of CKD patients as well as over-
growth of Proteobacteria,Actinobacteria,andFirmicutes in
the colon [49]. In particular, an expansion of bacterial species
with urease, uricase, and indole- and p-cresol-forming en-
zymes has been documented [60]. Urea is now considered a
key factor in the pathogenesis of increased permeability of the
Tabl e 3 Effects of chronic
kidney disease on intestinal
bacteria metabolism
Effects Mechanism
1.Reduced intake of dietary fibers Prescribed potassium restriction leads to reduced intake of fruits
and vegetables
2.Prolonged colonic transit time
(constipation)
Multifactorial: dialysis modality, lifestyle, inactivity, phosphate
binders, dietary restrictions, low fluid intake, primary renal
disease, and comorbidities (diabetes, heart failure, malnutrition,
cerebrovascular disease)
3.Increased amounts of protein available
for proteolytic bacterial species
Protein assimilation is impaired in uremia, with increased amounts
of intact proteins reaching the colon
4.Changes of the colonic microbiota Increased blood ammonia concentrations may change intestinal
lumen pH; drug therapies (antibiotics, phosphate binders,
antimetabolites etc.) with local effect in the gut lumen
5.Increased permeability of the intestinal
barrier
Uremia; hypervolemia and intestinal ischemia caused by
aggressive ultrafiltration volumes or intradialytic hypotension
From: Sabatino A et al. Nephrol Dial Transplant, 2015, 30:924933, by permission of Oxford University Press
[32]
16 Page 4 of 9 Curr Diab Rep (2017) 17:16
intestinal barrier. The increased influx of urea into the intesti-
nal lumen as a consequence of uremia may foster bacterial
species that produce urease. These bacterial species hydrolyze
urea to ammonia and increase intestinal pH, leading to muco-
sal irritation and structural damage [61,62]. In addition, intes-
tinal excretion of both uric acid and oxalate is also increased in
CKD [63,64]. Because of the wide availability of nitrogen
waste products in the intestine, the overgrowth of microbes
capable of utilizing these substrates is thus favored [60].
CKD is characterized by the progressive accumulation of
many substances and solutes, such as electrolytes, hormones,
and other solutes. Some of these compounds, termed uremic
toxins, may interfere with many biological functions and may
have important effects on inflammatory status and CVD risk [7,
65,66]. The identity of such toxins remains an active area of study
[67,68]. Their precursors are formed in the GI tract during bac-
terial protein fermentation. The two most widely studied of these
compounds are p-cresol (the precursor of p-cresyl-sulfate and p-
cresyl-glucuronide) and indole (the precursor of indoxyl sulfate),
generated respectively from the fermentation of amino acids ty-
rosine and tryptophan. In healthy subjects, the kidney excretes
these molecules by active tubular secretion, while in CKD in-
creased blood concentration of p-cresyl-sulfate and indoxyl sul-
fate follows the reduction of renal function [69]. In ESRD patients
on dialysis, the clearance of p-cresyl-sulfate and indoxyl sulfate is
less than 10% of that of healthy subjects [70], and their increased
blood concentration correlates with poor outcomes [7177].
These protein-bound compounds negatively affect endothelial
function and repair by several mechanisms, including inflamma-
tion, oxidative stress, impaired nitric oxide production, and inhi-
bition of endothelial proliferation and healing [7378].
Targeting the Intestinal Microbiota to Modulate
Intestinal Barrier Dysfunction and Dysbiosis
in T2DM and CKD/ESRD
Probiotics and Prebiotics in T2DM
Probiotics are viable organisms that, when ingested in sufficient
amounts, exert positive health effects[79]. Among the many
claimed health benefits associated with probiotic bacteria, partic-
ularly important is the (transient) modulation of the intestinal
microbiota and the capability to interact with the immune system.
The term prebiotic refers to a selectively fermented ingredient
that allows specific changes, both in the composition and/or ac-
tivity in the gastrointestinal microbiota that confer benefits[80].
Prebiotics promote the growth of bacterial species that stabilize
the mucosal barrier function, reduce the abundance of pathogenic
bacteria by intestinal lumen acidification, overcome the compe-
tition for nutrients, and produce antimicrobial substances [81].
Some characteristics must be present in the food ingredient to be
classified as a prebiotic, such as resistance to digestion and
absorption in the upper gastrointestinal tract, easy fermentability
by the intestinal microbiota, and capability of selective stimula-
tion of growth and/or activity of beneficial bacteria potentially
associated with health and well-being [79,80]. Finally, when
probiotics are administered along with prebiotics, the combina-
tion is referred to as synbiotics.
It remains to be fully ascertained whether probiotic admin-
istration may represent an efficacious treatment for T2DM.
Recent literature has focused on targeting bacterial strains
and increased intestinal permeability by SCFAs. SCFAs have
been shown to modulate intestinal hormones and to have im-
portant effects in metabolic health since they affect intestinal
permeability, satiety, gastric emptying, and food intake [82,
83]. It appears that butyrate plays a pivotal role in the correc-
tion of endotoxemia, by improving intestinal wall barrier
function, with proliferation of colonic epithelial cells and
tight-junction tightness [84]. In a landmark study performed
in 18 obese subjects with metabolic syndrome and insulin
resistance, fecal transplantation from lean donors, but not au-
tologous transplantation, improved insulin sensitivity and in-
creased the microbiota diversity, in particular butyrate-
producing bacteria such as Roseburia intestinalis,
Faecalibacterium prausnitzii, and Eubacterium hallii [85].
There is also evidence that classical probiotics can stop weight
gain and improve glucose tolerance in mice with T2DM [86].
Research focusing on the intestinal barrier function demon-
strated that supplementation with 2 × 10
8
CFU/day of
Akkermansia muciniphila, a bacterium found in the mucus layer
of the intestinal wall, reduced serum LPS in mice fed a high-fat
diet [87]. However, data on A. muciniphila are still controversial
since some studies reported increased concentrations of this bac-
terial strain in some disease states or during high-fat diet [13,88].
Regarding the use of prebiotics, this approach was associ-
ated with favorable changes in the microbiota and improve-
ment of metabolic markers of obese mice [89]. In humans,
administration of prebiotics improved insulin sensitivity in
subjects without T2DM [90]. However, not all humans re-
spond to prebiotic treatment in the same manner, and lower
bacterial diversity is related to no response [91,92]. More
specifically, in a recent study, the Prevotella/Bacteroides ratio
of the intestinal microbiota of healthy subjects allowed the
identification of responders and non-responders [92]. In this
study, glucose metabolism of germ-free mice that received the
microbiota of responder subjects improved, while nothing
happened to germ-free mice that received the microbiota of
non-responders. More studies are needed to assess the role of
prebiotics and probiotics in the treatment of T2DM.
Probiotics and Prebiotics in CKD and ESRD
Currently, nutritional strategies aimed to modulate intestinal mi-
crobiota in CKD and to reduce the serum levels of uremic toxins
p-cresol and indoxyl sulfate are a promising area of research
Curr Diab Rep (2017) 17:16 Page 5 of 9 16
[93102]. Preliminary data already demonstrated the ability of a
functional food rich in prebiotic fibers (barley beta-glucans) to
modulate intestinal microbiota composition and metabolome in a
clinical trial involving healthy volunteers [103]. Moreover, beta-
glucans were able to increase fecal SCFA levels [103]andto
reduce circulating p-cresyl sulfate levels [104], demonstrating
their ability to induce a shift toward an intestinal metabolism
driven by saccharolytic bacteria.
In the context of CKD, several studies tried to modulate the
intestinal environment and microbiota by using probiotics
(i.e., Lactobacillus,Streptococcus,Bifidobacteria)[9396,
105], prebiotics (i.e., arabic gum, oligofructose) [9799], or
synbiotics (i.e., Lactobacillus and Bifidobacterium combined
with oligosaccharides) [101,102]. In most cases, prebiotics
and probiotics were administered to explore their effects on
blood accumulation of blood urea nitrogen (BUN), p-cresyl
sulfate, and/or indoxyl sulfate, which are the metabolic
byproducts of nitrogen-containing compounds. In one study
on probiotics, the use of a mix of bacteria (Lactobacillus
acidophilus KB27, Bifidobacterium longum KB31, and
Streptococcus thermophilus KB19) for 6 months reduced
BUN and uric acid levels in stage 34 CKD patients [94]. In
amorerecentstudy[96], a 2-month treatment with a dairy
product containing 16 × 10
9
CFU of Lactobacillus casei
Shirota was able to reduce BUN concentration in CKD pa-
tients stages 3 and 4. Non-randomized studies on hemodialy-
sis (HD) patients [93,105] demonstrated a reduced excretion
of p-cresol and indican (i.e., a precursor of indoxyl sulfate)
and decreased serum levels of indoxyl sulfate [93,105], prob-
ably owing to reduced intestinal production of these toxins. In
CKD patients, the use of prebiotics also presented beneficial
effects, such as BUN decrease [98], improved eGFR [99],
higher fecal nitrogen excretion, and increased fecal
saccharolytic bacteria [89]. These data suggest that the pres-
ence of prebiotics provides enough energy substrates for the
intestinal microbiota, allowing saccharolytic bacteria to incor-
porate nitrogen for growth, thus reducing the production of
uremic compounds. Other studies have shown a reduction of
serum p-cresyl sulfate and p-cresyl sulfate generation rates in
ESRD patients on hemodialysis when patients received prebi-
otics [98] or fiber-enriched food [99]. The use of synbiotics
decreased serum p-cresol conjugate levels [100102], normal-
ized the amount and consistency of stool in HD patients [100],
increased the counts of Bifidobacteria [101], and modified the
stool microbiome of HD patients [102]. However, studies in-
vestigating the impact of probiotics on clinical endpoints (i.e.,
CVD and mortality) are lacking.
Conclusion
There is increased interest in the complex and bidirectional
relationship between the host and its microbiota, especially
regarding the role on the development of non-communicable
disease such as obesity and diabetes. Since many studies are
cross-sectional, it is not possible at the present time to estab-
lish a clear-cut causal relationship between intestinal dysbiosis
and obesity or T2DM. However, we know that dysbiosis may
cause endotoxemia and chronic inflammation, both through
directly disrupting the intestinal barrier and by reducing the
number of beneficial bacteria that produce SCFAs. T2DM and
CKD, whose coexistence is increasingly found in clinical
practice, share similar negative effects on both intestinal mi-
crobiota and the intestine itself. Thus, prospective studies are
necessary to define causality, and further randomized con-
trolled trials are needed in both T2DM and CKD to fully
define the role of probiotic and prebiotic therapies.
Compliance with Ethical Standards
Conflict of Interest A.S., G.R., C.C., L.G., and E.F. declare that they
have no competing interests.
Human and Animal Rights and Informed Consent This article con-
tains studies with human subjects performed by one of the authors, L.G.
In this case, all procedures performed in studies involving human partic-
ipants were in accordance with the ethical standards of the institutional
research committee and with the Helsinki declaration. This article does
not contain any studies with animal subjects performed by any of the
authors.
References
Papers of interest, published recently, have been highlighted
as:
Of importance
•• Of major importance
1. WHO. Global status report on noncommunicable diseases 2014.
World Health Organization. 2014; 176.
2. Johnson AMF, Olefsky JM. The origins and drivers of insulin
resistance. Cell. 2013;152:67384.
3. Tilg H, Moschen A. Inflammatory mechanisms in the regulation
of insulin resistance. Mol Med. 2008;14:22231.
4. American Diabetes Association. National Diabetes Statistics
Report: 2014 Estimates of Diabetes and Its Burden in the
Epidemiologic estimation methods. Natl Diabetes Stat Rep.
2014; 20092012.
5. Gansevoort RT, Correa-Rotter R, Hemmelgarn BR, Jafar TH,
Heerspink HJL, Mann JF, et al. Chronic kidney disease and car-
diovascular risk: epidemiology, mechanisms, and prevention.
Lancet. 2013;382:33952.
6. 2013 USRDS Annual Data Report: Atlas of Chronic Kidney
Disease and End-Stage Renal Disease in the United States. Am J
Kidney Dis 2014; 63: supplement e1e478.
7. Carrero JJ, Stenvinkel P. Persistent inflammation as a catalyst for
other risk factors in chronic kidney disease: a hypothesis proposal.
Clin J Am Soc Nephrol. 2009;4:S4955.
8. Stenvinkel P, Carrero JJ, Axelsson J, Lindholm B, Heimburger O,
Massy Z. Emerging biomarkers for evaluating cardiovascular risk
16 Page 6 of 9 Curr Diab Rep (2017) 17:16
in the chronic kidney disease patient: how do new pieces fit into
the uremic puzzle? Clin J Am Soc Nephrol. 2008;3:50521.
9. Honda H, Qureshi AR, Heimburger O, Barany P, Wang K, et al.
Serum albumin, C-reactive protein, interleukin-6, and fetuin A as
predictors of malnutrition, cardiovascular disease, and mortality in
patients with ESRD. Am J Kidney Dis. 2006;47:13948.
10. Carrero JJ, Yilmaz MI, Lindholm B, Stevinkel P. Cytokine dys-
regulation in chronic kidney disease: how can we treat it? Blood
Purif. 2008;26:2919.
11. Le Chatelier E, Nielsen T, Qin J, Prifti E, Hildebrand F, Falony G,
et al. Richness of human gut microbiome correlates with metabolic
markers. Nature. 2013;500:5416.
12. Larsen N, Vogensen FK, Van Den Berg FWJ, Nielsen DS,
Andreasen AS, Pedersen BK, et al. Gut microbiota in human
adults with type 2 diabetes differs from non-diabetic adults.
PLoS One. 2010;5:e9085.
13. Wang J, Qin J, Li Y, Cai Z, Li S, Zhu J, et al. A metagenome-wide
association study of gut microbiota in type 2 diabetes. Nature.
2012;490:5560.
14. Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D,
et al. Metabolic endotoxemia initiates obesity and insulin resis-
tance. Diabetes. 2007;56:176172.
15. Erridge C, Attina T, Spickett CM, Webb DJ. A high-fat meal
induces low-grade endotoxemia: evidence of a novel mechanism
of postprandial inflammation. Am J Clin Nutr. 2007;86:128692.
16. Harte AL, Varma MC, Tripathi G, Mcgee KC, Al-Daghri NM, Al-
Attas OS, et al. High fat intake leads to acute postprandial expo-
sure to circulating endotoxin in type 2 diabetic subjects. Diabetes
Care. 2012;35:37582.
17. Hildebrandt M, Hoffman C, Sherrill-mix S, Keilbaugh S, Chen Y,
Knight R, et al. High-fat diet determines the composition of the
murine gut microbiome independently of the obesity.
Gastroenterology. 2009;137:171624.
18. Stenvinkel P. Inflammation in end-stage renal failure: could it be
treated? Nephrol Dial Transpl. 2002;17:S338.
19. Kalantar-Zadeh K, Brennan ML, Hazen SL. Serum
myeloperoxidase and mortality in maintenance hemodialysis pa-
tients. Am J Kidney Dis. 2006;48:5968.
20. Moller DE, Kaufman KD. Metabolic syndrome: a clinical and
molecular perspective. Annu Rev Med. 2005;56:4562.
21. Kau AL, Ahern PP, Griffin NW, Goodman AL, Jeffrey I. Human
nutrition, the gut microbiome, and immune system: envisioning
the future. Nature. 2012;474:32736.
22. Carrero JJ, Stenvinkel P, Cuppari L, Ikizler TA, Kalantar-Zadeh K,
Kaysen G, et al. Etiology of the protein-energy wasting syndrome
in chronic kidney disease: a consensus statement from the
International Society of Renal Nutrition and Metabolism
(ISRNM). J Ren Nutr. 2013;23:7790.
23. Cerf-Bensussan N, Eberl G. The dialog between microbiota and
the immune system: shaping the partners through development
and evolution. Semin Immunol. 2012;24:12.
24. Szeto CC, Kwan BC, Chow KM, Lai KB, Chung KY, Leung CB,
et al. Endotoxemia Is related to systemic inflammation and ath-
erosclerosis in peritoneal dialysis patients. Clin J Am Soc Nephrol.
2008;3:4316.
25. Gonçalves S, Pecoits-Filho R, Perreto S, Barberato SH, Stinghen
AEM, Lima EGA, et al. Associations between renal function,
volume status and endotoxaemia in chronic kidney disease pa-
tients. Nephrol Dial Transplant. 2006;21:278894.
26. Anders H-J, Andersen K, Stecher B. The intestinal microbiota, a
leaky gut, and abnormal immunity in kidney disease. Kidney Int.
2013;83:10106.
27. Evenepoel P, Meijers BKI, Bammens BRM, Verbeke K. Uremic
toxins originating from colonic microbial metabolism. Kidney Int.
2009;76:S129.
28.Aron-Wisnewsky J, Clément K. The gut microbiome, diet and
links to cardiometabolic and chronic disorders. Nat Rev
Nephrol. 2016;12:16981. This review is important because
summarizes data suggesting a link between the intestinal mi-
crobiota and derived metabolites with food intake patterns,
metabolic alterations and chronic cardiometabolic diseases.
29. Pflughoeft KJ, Versalovic J. Human microbiome in health and
disease. Annu Rev Pathol. 2012;7:99122.
30. Human Microbiome Project Consortium. Structure, function and
diversity of the healthy human microbiome. Nature. 2012;486:
20714.
31. Hollister EB, Gao C, Versalovic J. Compositional and functional
features of the gastrointestinal microbiome and their effects on
human health. Gastroenterology. 2014;146:144958.
32.Sabatino A, Regolisti G, Brusasco I, Cabassi A, Morabito S,
Fiaccadori E. Alterations of intestinal barrier and microbiota in
chronic kidney disease. Nephrol Dial Transplant. 2015;30:924
33. This review is relevant because it discusses in depth the
cross-talk between the kidney and the intestine in CKD and
ESRD.
33.•• Vaziri ND, Zhao YY, Pahl MV. Altered intestinal microbial flora
and impaired epithelial barrier structure and function in CKD: the
nature, mechanisms, consequences and potential treatment.
Nephrol Dial Transplant. 2016;31:73746. This review gives a
thorough description on the mechanisms involved in the de-
rangement of intestinal barrier in CKD and ESRD.
34. Baumgart DC, Dignass U. Intestinal barrier function. Curr Opin
Clin Nutr Metab Care. 2002;5:68594.
35. Gregor MF, Hotamisligil GS. Inflammatory mechanisms in obe-
sity. Annu Rev Immunol. 2011;29:41545.
36. Ley RE, Bäckhed F, Turnbaugh P, Lozupone CA, Knight RD,
Gordon JI. Obesity alters gut microbial ecology. Proc Natl Acad
Sci U S A. 2005;102:110705.
37. Turnbaugh PJ, Bäckhed F, Fulton L, Gordon JI. Diet-induced
obesity is linked to marked but reversible alterations in the mouse
distal gut microbiome. Cell Host Microbe. 2008;3:21323.
38. Bäckhed F, Manchester JK, Semenkovich CF, Gordon JI.
Mechanisms underlying the resistance to diet-induced obesity in
germ-free mice. Proc Natl Acad Sci. 2007;104:97984.
39. Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE, Kau L, et al.
Gut microbiota from twins discordant for obesity modulate me-
tabolism in mice gut microbiota from twins metabolism in mice.
Science. 2013;341:1241214.
40. Karlsson FH, Tremaroli V, Nookaew I, Bergström G, Behre CJ,
Fagerberg B, et al. Gut metagenome in European women with
normal, impaired and diabetic glucose control. Nature.
2013;498:99103.
41. Ley R, Turnbaugh P, Klein S, Gordon J. Microbial ecology: hu-
man gut microbes associated with obesity. Nature. 2006;444:
10223.
42. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER,
Gordon JI. An obesity-associated gut microbiome with increased
capacity for energy harvest. Nature. 2006;444:102731.
43. Gao Z, Yin J, Zhang J, Ward RE, Martin RJ, Lefevre M, et al.
Butyrate improves insulin sensitivity and increases energy expen-
diture in mice. Diabetes. 2009;58:150917.
44. Lin HV, Frassetto A, Kowalik EJ, Nawrocki AR, Lu MM,
Kosinski JR, et al. Butyrate and propionate protect against diet-
induced obesity and regulate gut hormones via free fatty acid
receptor 3-independent mechanisms. PLoS One. 2012;7:e35240.
45. Wichmann A, Allahyar A, Greiner TU, Plovier H, Lundén GO,
Larsson T, et al. Microbial modulation of energy availability in the
colon regulates intestinal transit. Cell Host Microbe. 2013;14:
58290.
46. Vallon V, Docherty NG. Intestinal regulation of urinary sodium
excretion and the pathophysiology of diabetic kidney disease: a
Curr Diab Rep (2017) 17:16 Page 7 of 9 16
focus on glucagon-like peptide 1 and dipeptidyl peptidase 4 in
diabetic kidney disease. Exp Physiol. 2014;99:11405.
47. Allin KH, Nielsen T, Pedersen O. Mechanisms in endocrinology:
gut microbiota in patients with type 2 diabetes mellitus. Eur J
Endocrinol. 2015;172:16777.
48. Ramezani A, Massy ZA, Meijers B, Evenepoel P, Vanholder R,
Raj DS. Role of the gut microbiome in uremia: a potential thera-
peutic target. Am J Kidney Dis. 2016;67:48398.
49. Vaziri ND, Wong J, Pahl M, Piceno YM, Yuan J, DeSantis TZ,
et al. Chronic kidney disease alters intestinal microbial flora.
Kidney Int. 2012;83:30815.
50. De Angelis M, Montemurno E, Piccolo M, Vannini L, Lauriero G,
Maranzano V, et al. Microbiota and metabolome associated with
immunoglobulin a nephropathy (IgAN). PLoS One. 2014;9:
e99006.
51. Piccolo M, De Angelis M, Lauriero G, Montemurno E, Di Cagno
R, Gesualdo L, et al. Salivary microbiota associated with immu-
noglobulin A nephropathy. Microb Ecol. 2015;70:55765.
52. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button
JE, Wolfe BE, et al. Diet rapidly and reproducibly alters the human
gut microbiome. Nature. 2014;505:55963.
53. Krishnamurthy VMR, Wei G, Baird BC, Murtaugh M, Chonchol
MB, Raphael KL, et al. High dietary fiberintake is associated with
decreased inflammation and all-cause mortality in patients with
chronic kidney disease. Kidney Int. 2012;81:3006.
54. Bammens B, Verbeke K, Vanrenterghem Y, Evenepoel P.
Evidence for impaired assimilation of protein in chronic renal
failure. Kidney Int. 2003;64:2196203.
55. Mafra D, Barros AF, Fouque D. Dietary protein metabolism by gut
microbiota and its consequences for chronic kidney disease pa-
tients. Futur Microbiol. 2013;8:131723.
56. Nyangale EP, Mottram DS, Gibson GR. Gut microbial activity,
implications for health and disease: the potential role of metabolite
analysis. J Proteome Res. 2012;11:557385.
57. Bammens B, Evenepoel P, Verbeke K, Vanrenterghem Y.
Impairment of small intestinal protein assimilation in patients with
end-stage renal disease: extending the malnutritioninflamma-
tionatherosclerosis concept. Am J Clin Nutr. 2004;80:153643.
58. Montemurno E, Cosola C, Dalfino G, Daidone G, De Angelis M,
Gobbetti M, et al. What would you like to eat, Mr CKD
Microbiota? A Mediterranean Diet, please! Kidney Blood Press
Res. 2014;39:11423.
59. Barrios C, Beaumont M, Pallister T, Villar J, Goodrich JK, Clark
A, et al. Gutmicrobiotametabolite axis in early renal function
decline. PLoS One. 2015;10:e0134311.
60. Wong J, Piceno YM, DeSantis TZ, Pahl M, Andersen GL, Vaziri
ND. Expansion of urease- and uricase-containing, indole- and p-
cresol-forming and contraction of short-chain fatty acid-producing
intestinal microbiota in ESRD. Am J Nephrol. 2014;39:2307.
61. Kang JY. The gastrointestinal tract in uremia. Dig Dis Sci.
1993;38:25768.
62. Vaziri N, Freel W, Hatch M. Effect of chronic experimental renal
insufficiency on urate metabolism. J Am Soc Nephrol. 1995;6:
13137.
63. Hatch M, Vaziri ND. Enhanced enteric excretion of urate in rats
with chronic renal failure. Clin Sci (Lond). 1994;86:5116.
64. Hatch M, Freel RW, Vaziri ND. Intestinal excretion of oxalate in
chronic renal failure. J Am Soc Nephrol. 1994;5:133943.
65. Neirynck N, Vanholder R, Schepers E, Eloot S, Pletinck A,
Glorieux G. An update on uremic toxins. Int Urol Nephrol.
2013;45:13950.
66. Sirich TL, Funk BA, Plummer NS, Hostetter TH, Meyer TW.
Prominent accumulation in hemodialysis patients of solutes nor-
mally cleared by tubular secretion. J Am Soc Nephrol. 2014;25:
61522.
67. Vanholder R, Meert N, Schepers E, Glorieux G. Uremic toxins: do
we know enough to explain uremia? Blood Purif. 2008;26:7781.
68. Vanholder R, Baurmeister U, Brunet P, Cohen G, Glorieux G,
Jankowski J, et al. A bench to bedside view of uremic toxins. J
Am Soc Nephrol. 2008;19:86370.
69. Viaene L, Meijers BKI, Bammens B, Vanrenterghem Y,
Evenepoel P. Serum concentrations of p-cresyl sulfate and indox-
yl sulfate, but not inflammatory markers, increase in incident
peritoneal dialysis patients in parallel with loss of residual renal
function. Perit Dial Int. 2014;34:718.
70. Dobre M, Meyer TW, Hostetter TH. Searching for uremic toxins.
Clin J Am Soc Nephrol. 2013;8:3227.
71. Meijers BKI, Claes K, Bammens B, De Loor H, Viaene L,
Verbeke K, et al. p-Cresol and cardiovascular risk in mild-to-
moderate kidney disease. Clin J Am Soc Nephrol. 2010;5:11829.
72. Liabeuf S, Barreto DV, Barreto FC, Meert N, Glorieux G,
Schepers E, et al. Free p-cresylsulphate is a predictor of mortality
in patients at different stages of chronic kidney disease. Nephrol
Dial Transplant. 2010;25:118391.
73. Jourde-Chiche N, Dou L, Cerini C, Dignat-George F, Brunet P.
Vascular incompetence in dialysis patientsprotein-bound ure-
mic toxins and endothelial dysfunction. Semin Dial. 2011;24:
32737.
74. Ito S, Yoshida M. Protein-bound uremic toxins: new culprits of
cardiovascular events in chronic kidney disease patients. Toxins
(Basel). 2014;6:66578.
75. Dou L, Jourde-Chiche N, Faure V, Cerini C, Berland Y, Dignat-
George F, et al. The uremic solute indoxyl sulfate induces oxida-
tive stress in endothelial cells. J Thromb Haemost. 2007;5:1302
8.
76. Yu M, Kim YJ, Kang D-H. Indoxyl sulfate-induced endothelial
dysfunction in patients with chronic kidney disease via an induc-
tion of oxidative stress. Clin J Am Soc Nephrol. 2011;6:309.
77. Barreto FC, Barreto DV, Liabeuf S, Meert N, Glorieux G, Temmar
M, et al. Serum indoxyl sulfate is associated with vascular disease
and mortality in chronic kidney disease patients. Clin J Am Soc
Nephrol. 2009;4:15518.
78. Rossi M, Campbell KL, Johnson DW, Stanton T, Vesey DA,
Coombes JS, et al. Protein-bound uremic toxins, inflammation
and oxidative stress: a cross-sectional study in stage 34 chronic
kidney disease. Arch Med Res. 2014;45:30917.
79. Chow J. Probiotics and prebiotics: a brief overview. J Ren Nutr.
2002;12:7686.
80. de Vrese M, Schrezenmeir J. Probiotics, prebiotics and synbiotics.
Adv Biochem Eng Biotechnol. 2008;111:166.
81. Griffiths EA, Duffy LC, Schanbacher FL, Qiao H, Dryja D,
Leavens A, et al. In vivo effects of bifidobacteria and lactoferrin
on gut endotoxin concentration and mucosal immunity in Balb/c
mice. Dig Dis Sci. 2004;49:57989.
82. Lovshin JA, Drucker DJ. Incretin-based therapies for type 2 dia-
betes mellitus. Nat Rev Endocrinol. 2009;5:2629.
83. Veldhorst MAB, Westerterp KR, Westerterp-Plantenga MS.
Gluconeogenesis and protein-induced satiety. Br J Nutr.
2012;107:595600.
84. Monolayers C-C, Peng L, Li Z, Green RS, Holzman IR, Lin J.
Butyrate enhances the intestinal barrier by facilitating tight junc-
tion assembly via activation of AMP-activated protein kinase. J
Nutr. 2009;139:161925.
85. Vrieze A, Van Nood E, Holleman F, Salojarvi J, Kootte RS,
Bartelsman JF, et al. Transfer of intestinal microbiota from lean
donors increases insulin sensitivity in individuals with metabolic
syndrome. Gastroenterology. 2012;143:913916.e7.
86. Yadav H, Lee JH, Lloyd J, Walter P, Rane SG. Beneficial meta-
bolic effects of a probiotic via butyrate-induced GLP-1 hormone
secretion. J Biol Chem. 2013;288:2508897.
16 Page 8 of 9 Curr Diab Rep (2017) 17:16
87. Everard A, Belzer C, Geurts L, Ouwerkerk JP, Druart C, Bindels
LB, et al. Cross-talk between Akkermansia muciniphila and intes-
tinal epithelium controls diet-induced obesity. Proc Natl Acad Sci
U S A. 2013;110:906671.
88. Jakobsdottir G, Xu J, Molin G, Ahrne S, Nyman M. High-fat diet
reduces the formation of butyrate, but increases succinate, inflam-
mation, liver fat and cholesterol in rats, while dietary fibre coun-
teracts these effects. PLoS One. 2013;8:e80476.
89. Cani PD, Neyrinck AM, Fava F, Knauf C, Burcelin RG, Tuohy
KM, et al. Selective increases of bifidobacteria in gut microflora
improve high-fat-diet-induced diabetes in mice through a mecha-
nism associated with endotoxaemia. Diabetologia. 2007;50:2374
83.
90. Robertson MD, Bickerton AS, Dennis AL, Vidal H, Frayn KN.
Insulin-sensitizing effects of dietary resistant starch and effects on
skeletal muscle and adipose tissue metabolism. Am J Clin Nutr.
2005;82:55967.
91. Salonen A, Lahti L, Salojarvi J, Holtrop G, Korpela K, Duncan
SH, et al. Impact of diet and individual variation on intestinal
microbiota composition and fermentation products in obese men.
ISME J. 2014;8:221830.
92. Kovatcheva-Datchary P, Nilsson A, Akrami R, Lee YS, De
Vadder F, Arora T, et al. Dietary fiber-induced improvement in
glucose metabolism Is associated with increased abundance of
Prevotella. Cell Metab. 2015;22:97182.
93. Takayama F, Taki K, Niwa T. Bifidobacterium in gastro-resistant
seamless capsule reduces serum levels of indoxyl sulfate in pa-
tients on hemodialysis. Am J Kidney Dis. 2003;41:S1425.
94. Ranganathan N, Friedman EA, Tam P, Rao V, Ranganathan P,
Dheer R. Probiotic dietary supplementation in patients with stage
3 and 4 chronic kidney disease: a 6-month pilot scale trial in
Canada. Curr Med Res Opin. 2009;25:191930.
95. Ranganathan N, Ranganathan P, Friedman EA, Joseph A, Delano
B, Goldfarb DS, et al. Pilot study of probiotic dietary supplemen-
tation for promoting healthy kidney function in patients with
chronic kidney disease. Adv Ther. 2010;27:63447.
96. Miranda Alatriste PV, Urbina Arronte R, Gomez Espinosa CO, de
los Espinosa Cuevas M. Effect of probiotics on human blood urea
levels in patients with chronic renal failure. Nutr Hosp. 2014;29:
58290.
97. Bliss DZ, Stein TP, Schleifer CR, Settle RG. Supplementation
with gum arabic fiber increases fecal nitrogen excretion and
lowers serum urea nitrogen concentration in chronic renal failure
patients consuming a low-protein diet. Am J Clin Nutr. 1996;63:
3928.
98. Meijers BKI, De Preter V, Verbeke K, Vanrenterghem Y,
Evenepoel P. p-Cresyl sulfate serum concentrations in
haemodialysis patients are reduced by the prebiotic
oligofructose-enriched inulin. Nephrol Dial Transplant. 2010;25:
21924.
99. Salmean YA, Segal MS, Langkamp-Henken B, Canales MT, Zello
GA, Dahl WJ. Foods with added fiber lower serum creatinine
levels in patients with chronic kidney disease. J Ren Nutr.
2013;23:e2932.
100. Nakabayashi I, Nakamura M, Kawakami K, Ohta T, Kato I,
Uchida K, et al. Effects of synbiotic treatment on serum level of
p-cresol in haemodialysis patients: a preliminary study. Nephrol
Dial Transplant. 2011;26:10948.
101. Cruz-Mora J, Martínez-Hernández NE, Martín del Campo-López
F, Viramontes-Hörner D, Vizmanos-Lamotte B, Muñoz-Valle JF,
et al. Effects of a symbiotic on gut microbiota in Mexican patients
with end-stage renal disease. J Ren Nutr. 2014;24:3305.
102. Rossi M, Johnson DW, Morrison M, Pascoe EM, Coombes JS,
Forbes JM, et al. Synbiotics easing renal failure by improving gut
microbiology (SYNERGY): a randomized trial. Clin J Am Soc
Nephrol. 2016;11:22331.
103. De Angelis M, Montemurno E, Vannini L, Cosola C, Cavallo N,
Gozzi G, et al. Effect of whole-grain barley on the human fecal
microbiota and metabolome. Appl Environ Microbiol. 2015;81:
794556.
104. Cosola C, De Angelis M, Rocchetti MT, Montemurno E,
Maranzano V, Dalfino G, et al. Beta-glucans supplementation
associates with reduction in p-cresyl sulfate levels and improved
endothelial vascular reactivity in healthy individuals. PLoS One.
2017;12(1):e0169635. doi:10.1371/journal.pone.0169635.
105. Hida M, Aiba Y, Sawamura S, Suzuki N, Satoh T, Koga Y.
Inhibition of the accumulation of uremic toxins in the blood and
their precursors in the feces after oral administration of Lebenin, a
lactic acid bacteria preparation, to uremic patients undergoing he-
modialysis. Nephron. 1996;74:34955.
Curr Diab Rep (2017) 17:16 Page 9 of 9 16
... To the best of our knowledge, the present study is one of few studies that has focused on gut microbiome alterations in T2DM and ESRD (32,33). There is ample information regarding the characteristics of the intestinal microbiome in T2DM, and in CKD and ESRD, respectively; however, not much data exists regarding patients suffering from both pathologies. ...
... Studies have already been performed that have focused on the alterations of the intestinal microbiome in both diabetes and CKD/ESRD, but there are very few, if any, studies that have focused on patients with this specific association of pathologies (32,33). Thus, the present results could shed further light upon the complex interrelations that appear between the two aforementioned pathologies and the gut microbiome. ...
Article
Full-text available
Chronic kidney disease (CKD) is a widespread health concern, which affects ~9.1% of the global population and 12-15% of individuals in upper-middle income countries. Notably, ~2% of patients with CKD progress to end-stage renal disease (ESRD), which leads to a substantial decline in the quality of life, an increased risk of mortality and significant financial burden. Patients with ESRD often still suffer from uremia and uremic syndromes, due to the accumulation of toxins between dialysis sessions and the inadequate removal of protein-bound toxins during dialysis. A number of these toxins are produced by the gut microbiota through the fermentation of dietary proteins or cholines. Furthermore, the gut microbial community serves a key role in maintaining metabolic and immune equilibrium in individuals. The present study aimed to investigate the gut microbiota patterns in individuals with type 2 diabetes mellitus (T2DM) and ESRD via quantitative PCR analysis of the 16S and 18S ribosomal RNA of selected members of the gut microbiota. Individuals affected by both T2DM and ESRD displayed distinctive features within their intestinal microbiota. Specifically, there were increased levels of Gammaproteobacteria observed in these patients, and all subjects exhibited a notably increased presence of Enterobacteriaceae compared with healthy individuals. This particular microbial community has established connections with the presence of inflammatory processes in the colon. Moreover, the elevated levels of Enterobacteriaceae may serve as an indicator of an imbalance in the intestinal microbiota, a condition known as dysbiosis. In addition, the Betaproteobacteria phylum was significantly more prevalent in the stool samples of patients with both T2DM and ESRD when compared with the control group. In conclusion, the present pilot study focused on gut microbiome alterations in T2DM and ESRD. Understanding the relationship between dysbiosis and CKD may identify new areas of research and therapeutic interventions aimed at modulating the gut microbiota to improve the health and outcomes of individuals with CKD and ESRD.
... Specifically, Eggerthella lenta and Fusobacterium nucleatum can accelerate the accumulation of phenylacetylglycine, phenyl sulphate, and indoxyl sulphate in the blood of CKD mouse models, leading to an increase in the severity of glomerulosclerosis and renal fibrosis [5]. Several studies have also established a correlation between gut bacteria and CKD clinical characteristics such as diabetes, proteinuria, elevated levels of inflammatory cytokines, and increasing galactose-deficient IgA1 [6][7][8]. ...
... In this case, the analysis includes another fungal species in addition to the two variates with the largest adjusted R 2 from the second PERMANOVA analysis. (6) The process continues until the largest adjusted R 2 from the last PERMANOVA analysis is smaller than that from the previous PERMANOVA analysis. The latter is then considered as the explanatory power of the gut mycobiome on the serum metabolome. ...
Article
Full-text available
Background The relationship between the gut mycobiome and end-stage renal disease (ESRD) remains largely unexplored. Methods In this study, we compared the gut fungal populations of 223 ESRD patients and 69 healthy controls (HCs) based on shotgun metagenomic sequencing data, and analyzed their associations with host serum and fecal metabolites. Results Our findings revealed that ESRD patients had a higher diversity in the gut mycobiome compared to HCs. Dysbiosis of the gut mycobiome in ESRD patients was characterized by a decrease of Saccharomyces cerevisiae and an increase in various opportunistic pathogens, such as Aspergillus fumigatus, Cladophialophora immunda, Exophiala spinifera, Hortaea werneckii, Trichophyton rubrum, and others. Through multi-omics analysis, we observed a substantial contribution of the gut mycobiome to host serum and fecal metabolomes. The opportunistic pathogens enriched in ESRD patients were frequently and positively correlated with the levels of creatinine, homocysteine, and phenylacetylglycine in the serum. The populations of Saccharomyces, including the HC-enriched Saccharomyces cerevisiae, were frequently and negatively correlated with the levels of various toxic metabolites in the feces. Conclusions Our results provided a comprehensive understanding of the associations between the gut mycobiome and the development of ESRD, which had important implications for guiding future therapeutic studies in this field.
... A significant pathophysiological mechanism that unites the diverse cardio-metabolic disease phenotypes is lowgrade chronic inflammation (Donath et al. 2019). Studies (Danesh 2000;Gabriel and Ferguson 2023) suggested that chronic systemic low-grade inflammation associated with CMD disorders may be partially explained by dysregulation in the microbial balance that results in unchecked immune activation; endotoxemia (production of lipopolysaccharides), disruption of the intestinal barrier, and loss of beneficial bacteria which aid in the CMD progression (Sabatino et al. 2017). Evidence through reported studies suggested reduced gut microbial diversity, altered relative abundances of main phyla Firmicutes and Bacteroidetes as well as low-grade inflammation have all been linked to risk factors of CMD, like Type 2 diabetes (T2D), obesity, and hypertension (Ley et al. 2006;Larsen et al. 2010;Cotillard et al. 2013;Li et al. 2017;Yan et al. 2017;Jama et al. 2019;Al-Reshed et al. 2023). ...
Article
Full-text available
Cardio-metabolic disease is a significant global health challenge with increasing prevalence. Recent research underscores the disruption of gut microbial balance as a key factor in disease susceptibility. We aimed to characterize the gut microbiota composition and function in cardio-metabolic disease and healthy controls. For this purpose, we collected stool samples of 18 subjects (12 diseased, 6 healthy) and we performed metagenomics analysis and functional prediction using QIIME2 and PICRUSt. Furthermore, we carried out assessments of microbe–gene interactions, gene ontology, and microbe–disease associations. Our findings revealed distinct microbial patterns in the diseased group, particularly evident in lower taxonomic levels with significant variations in 14 microbial features. The diseased cohort exhibited an enrichment of Lachnospiraceae family, correlating with obesity, insulin resistance, and metabolic disturbances. Conversely, reduced levels of Clostridium, Gemmiger, and Ruminococcus genera indicated a potential inflammatory state, linked to compromised butyrate production and gut permeability. Functional analyses highlighted dysregulated pathways in amino acid metabolism and energy equilibrium, with perturbations correlating with elevated branch-chain amino acid levels—a known contributor to insulin resistance and type 2 diabetes. These findings were consistent across biomarker assessments, microbe–gene associations, and gene ontology analyses, emphasizing the intricate interplay between gut microbial dysbiosis and cardio-metabolic disease progression. In conclusion, our study unveils significant shifts in gut microbial composition and function in cardio-metabolic disease, emphasizing the broader implications of microbial dysregulation. Addressing gut microbial balance emerges as a crucial therapeutic target in managing cardio-metabolic disease burden.
... Many studies have shown significant changes in the gut microbiota of patients with DN. Dysbiosis of the gut microbiota in DN patients is associated with endotoxemia, inflammation , intestinal barrier dysfunction (Xiong et al., 2019;Sun X. et al., 2022;Xu et al., 2022), and a decrease in beneficial bacteria that produce SCFAs (Sabatino et al., 2017). Pathogenic bacteria, such as Clostridium, Bacteroides, and Prevotella, can increase intestinal barrier permeability by producing toxins (Das et al., 2021). ...
Article
Full-text available
Diabetic nephropathy (DN) is one of the main complications of diabetes and a major cause of end-stage renal disease, which has a severe impact on the quality of life of patients. Strict control of blood sugar and blood pressure, including the use of renin–angiotensin–aldosterone system inhibitors, can delay the progression of diabetic nephropathy but cannot prevent it from eventually developing into end-stage renal disease. In recent years, many studies have shown a close relationship between gut microbiota imbalance and the occurrence and development of DN. This review discusses the latest research findings on the correlation between gut microbiota and microbial metabolites in DN, including the manifestations of the gut microbiota and microbial metabolites in DN patients, the application of the gut microbiota and microbial metabolites in the diagnosis of DN, their role in disease progression, and so on, to elucidate the role of the gut microbiota and microbial metabolites in the occurrence and prevention of DN and provide a theoretical basis and methods for clinical diagnosis and treatment.
... The immune-enhancing properties of SCFAs have been widely studied in mammals. However, most past studies have concentrated on treating diseases rather than investigating their ability to boost immunity in healthy subjects (Sabatino et al., 2017). Specifically, a research gap exists in how dietary SCFAs affect innate immunity through cellular and molecular pathways. ...
Article
Full-text available
The research examined the impact of adding potassium diformate (KDF) and sodium diformate (NDF) to the diet of Siberian sturgeon (Acipenser baerii) and how it influenced their antioxidant levels and immune responses. The fish (n = 315; 17.85 ± 0.2 g initial weight) were fed with seven diets for 56 days, including a control group (unsupplemented diet) and groups with varying levels of KDF (0.15, 0.3, and 0.5%) and NDF (0.15, 0.3, and 0.5%). After the feeding trial, the groups that consumed KDF or NDF demonstrated a dose-responsive elevation in serum superoxide dismutase, catalase, and glutathione peroxidase levels, along with an overall increase in total antioxidant capacity compared to the control group (P < 0.05). These groups also had lower malondialdehyde levels (P < 0.05) and improved immune functions, including higher levels of immunoglobulin M, total protein, albumin, globulin, the activities of lysozyme, respiratory burst, and alternative complement (P < 0.05). Also, the serum bactericidal activity experienced a significant increase (maximum in 0.5% NDF treatment) in the groups subjected to experimental feeding compared to the control group (P < 0.05). The transcription levels of interleukin-1 beta (IL-1β), interleukin 8 (IL-8), tumor necrosis factor-alpha (TNF-α), high mobility group box 1 (AbHMGB1), and high mobility group box 2 (AbHMGB2) genes in the head kidney were significantly upregulated in fish fed with different doses of KDF or NDF compared to the control group (P < 0.05). The most significant upregulation was observed in the group that received 0.5% NDF. Moreover, the experimental groups demonstrated a lower cumulative mortality rate of the fish following exposure to Aeromonas hydrophila AB005 infection compared to the control group (P < 0.05), with the lowest mortality rate observed in the 0.5% NDF or KDF group. Overall, the results suggest that dietary KDF or NDF, especially 0.5% NDF, are promising as environmentally friendly options and sustainable immunostimulants in Siberian sturgeon aquaculture.
... Genetic factors, dietary choices, and lifestyle also play pivotal roles in the progression of DN. Recent research has shed light on the relationship between dysregulated gut microbiota (GM) and the onset and progression of DN (4,5). ...
Article
Full-text available
Background The causal association between gut microbiota (GM) and the development of diabetic nephropathy (DN) remains uncertain. We sought to explore this potential association using two-sample Mendelian randomization (MR) analysis. Methods Genome-wide association study (GWAS) data for GM were obtained from the MiBioGen consortium. GWAS data for DN and related phenotypes were collected from the FinngenR9 and CKDGen databases. The inverse variance weighted (IVW) model was used as the primary analysis model, supplemented by various sensitivity analyses. Heterogeneity was assessed using Cochran’s Q test, while horizontal pleiotropy was evaluated through MR-Egger regression and the MR-PRESSO global test. Reverse MR analysis was conducted to identify any reverse causal effects. Results Our analysis identified twenty-five bacterial taxa that have a causal association with DN and its related phenotypes (p < 0.05). Among them, only the g_Eubacterium_coprostanoligenes_group showed a significant causal association with type 1 DN (p < Bonferroni-adjusted p-value). Our findings remained consistent regardless of the analytical approach used, with all methods indicating the same direction of effect. No evidence of heterogeneity or horizontal pleiotropy was observed. Reverse MR analysis did not reveal any causal associations. Conclusions This study established a causal association between specific GM and DN. Our findings contribute to current understanding of the role of GM in the development of DN, offering potential insights for the prevention and treatment strategies for this condition.
... Patients with type 2 diabetes have a microbiota that is characterized by a reduction in butyrate-producing bacteria, a moderate dysbiosis, an environment that is proinflammatory, a decrease in the expression of genes involved in vitamin synthesis, an increase in serum LPS levels, and an increased intestinal permeability [94]. However, the gut microbiota contributes to energy generation via anaerobic digestion of food components, which results in SCFA such as acetate, propionate, and butyrate. ...
Article
Full-text available
A global epidemiological shift has been observed in recent decades, characterized by an increase in age-related disorders, notably non-communicable chronic diseases, such as type 2 diabetes mellitus, cardiovascular and neurodegenerative diseases, and cancer. An appreciable causal link between changes in the gut microbiota and the onset of these maladies has been recognized, offering an avenue for effective management. Kefir, a probiotic-enriched fermented food, has gained significance in this setting due to its promising resource for the development of functional or value-added food formulations and its ability to reshape gut microbial composition. This has led to increasing commercial interest worldwide as it presents a natural beverage replete with health-promoting microbes and several bioactive compounds. Given the substantial role of the gut microbiota in human health and the etiology of several diseases, we conducted a comprehensive synthesis covering a total of 33 investigations involving experimental animal models, aimed to elucidate the regulatory influence of bioactive compounds present in kefir on gut microbiota and their potential in promoting optimal health. This review underscores the outstanding nutritional properties of kefir as a central repository of bioactive compounds encompassing micronutrients and amino acids and delineates their regulatory effects at deficient, adequate, and supra-nutritional intakes on the gut microbiota and their broader physiological consequences. Furthermore, an investigation of putative mechanisms that govern the regulatory effects of kefir on the gut microbiota and its connections with various human diseases was discussed, along with potential applications in the food industry.
Article
Full-text available
In today’s industrialized society food consumption has changed immensely toward heightened red meat intake and use of artificial sweeteners instead of grains and vegetables or sugar, respectively. These dietary changes affect public health in general through an increased incidence of metabolic diseases like diabetes and obesity, with a further elevated risk for cardiorenal complications. Research shows that high red meat intake and artificial sweeteners ingestion can alter the microbial composition and further intestinal wall barrier permeability allowing increased transmission of uremic toxins like p-cresyl sulfate, indoxyl sulfate, trimethylamine n-oxide and phenylacetylglutamine into the blood stream causing an array of pathophysiological effects especially as a strain on the kidneys, since they are responsible for clearing out the toxins. In this review, we address how the burden of the Western diet affects the gut microbiome in altering the microbial composition and increasing the gut permeability for uremic toxins and the detrimental effects thereof on early vascular aging, the kidney per se and the blood-brain barrier, in addition to the potential implications for dietary changes/interventions to preserve the health issues related to chronic diseases in future.
Article
The increasing prevalence of type 2 diabetes (T2D) is attributed to the rapid shift in our environment, particularly the modern lifestyle. Recent research has decoded the link between T2D and various chronic conditions with compromised gut health. Enhancing our understanding of gut health and actively maintaining a healthy gut can significantly impact overall well-being, including the management of T2D. Notably, recent key findings highlight the positive effects of incorporating probiotics, prebiotics (beneficial live organisms from diverse food sources), and increased fiber intake in promoting a beneficial balance of intestinal flora, thereby fortifying the gut. Research has demonstrated that probiotics, such as specific strains such as Lactobacillus and Bifidobacterium species, play a crucial role in supporting gut health and preventing the onset of T2D. Conversely, a diet high in saturated fats and processed sugars has been identified as a contributor to poor gut health, leading to a condition known as gut dysbiosis. Moreover, studies indicate that an imbalanced gut is a contributing factor to insulin resistance in individuals with T2D. It is noteworthy that certain factors, such as the use of anti-inflammatory drugs, antibiotics, and nonsteroidal medications, can significantly disrupt gut health and contribute to imbalances. This review emphasizes the importance of reinforcing gut health through the inclusion of specific probiotic strains and adopting a high-fiber, plant-based diet. The consumption of such a diet appears to be an effective and favorable strategy for improving intestinal microbiota and, consequently, overall health, with a specific focus on preventing T2D.
Article
Full-text available
Background: Resistant starch may modulate insulin sensitivity, although the precise mechanism of this action is unknown. Objective: We studied the effects of resistant starch on insulin sensitivity and tissue metabolism. Design: We used a 4-wk supplementation period with 30 g resistant starch/d, compared with placebo, in 10 healthy subjects and assessed the results by using arteriovenous difference methods. Results: When assessed by euglycemic-hyperinsulinemic clamp, insulin sensitivity was higher after resistant starch supplementation than after placebo treatment (9.7 and 8.5 × 10⁻² mg glucose · kg⁻¹ · min⁻¹ · (mU insulin/L)⁻¹, respectively; P = 0.03); insulin sensitivity during the meal tolerance test (MTT) was 33% higher (P = 0.05). Forearm muscle glucose clearance during the MTT was also higher after resistant starch supplementation (P = 0.03) despite lower insulin concentrations (P = 0.02); glucose clearance adjusted for insulin was 44% higher. Subcutaneous abdominal adipose tissue nonesterified fatty acid (NEFA; P = 0.02) and glycerol (P = 0.05) release were lower with resistant starch supplementation, although systemic NEFA concentrations were not significantly altered. Short-chain fatty acid concentrations (acetate and propionate) were higher during the MTT (P = 0.05 and 0.01, respectively), as was acetate uptake by adipose tissue (P = 0.03). Fasting plasma ghrelin concentrations were higher with resistant starch supplementation (2769 compared with 2062 pg/mL; P = 0.03), although postprandial suppression (40–44%) did not differ significantly. Measurements of gene expression in adipose tissue and muscle were uninformative, which suggests effects at a metabolic level. The resistant starch supplement was well tolerated. Conclusion: These results suggest that dietary supplementation with resistant starch has the potential to improve insulin sensitivity. Further studies in insulin-resistant persons are needed.
Article
Full-text available
Background Oat and barley beta-glucans are prebiotic fibers known for their cholesterol-lowering activity, but their action on the human gut microbiota metabolism is still under research. Although the induction of short-chain fatty acids (SCFA) following their ingestion has previously been reported, no study has investigated their effects on proteolytic uremic toxins p-cresyl sulfate (pCS) and indoxyl sulfate (IS) levels, while others have failed to demonstrate an effect on the endothelial function measured through flow-mediated dilation (FMD). Objective The aim of our study was to evaluate whether a nutritional intervention with a functional pasta enriched with beta-glucans could promote a saccharolytic shift on the gut microbial metabolism and improve FMD. Methods We carried out a pilot study on 26 healthy volunteers who underwent a 2-month dietary treatment including a daily administration of Granoro “Cuore Mio” pasta enriched with barley beta-glucans (3g/100g). Blood and urine routine parameters, serum pCS/IS and FMD were evaluated before and after the dietary treatment. Results The nutritional treatment significantly reduced LDL and total cholesterol, as expected. Moreover, following beta-glucans supplementation we observed a reduction of serum pCS levels and an increase of FMD, while IS serum levels remained unchanged. Conclusions We demonstrated that a beta-glucans dietary intervention in healthy volunteers correlates with a saccharolytic shift on the gut microbiota metabolism, as suggested by the decrease of pCS and the increase of SCFA, and associates with an improved endothelial reactivity. Our pilot study suggests, in addition to cholesterol, novel pCS-lowering properties of beta-glucans, worthy to be confirmed in large-scale trials and particularly in contexts where the reduction of the microbial-derived uremic toxin pCS is of critical importance, such as in chronic kidney disease.
Article
Full-text available
Cardiometabolic diseases (CMDs) have been associated with changes in the composition of the gut microbiota, with links between the host environment and microbiota identified in preclinical models. High-throughput sequencing technology has facilitated in-depth studies of the gut microbiota, bacterial-derived metabolites, and their association with CMDs. Such strategies have shown that patients with CMDs frequently exhibit enrichment or depletion of certain bacterial groups in their resident microbiota compared to healthy individuals. Furthermore, the ability to transfer resident gut microbiota from mice or humans into germ-free mouse models, or between human patients, has enabled researchers to characterize the causative role of the gut microbiota in CMDs. These approaches have helped identify that dietary intake of choline, which is metabolized by the gut microbiota, is associated with cardiovascular outcomes in mice and humans. Trimethylamine N-oxide (TMAO) - a metabolite derived from the gut microbiota - is also associated with poor cardiovascular outcomes in patients with cardiovascular disease and is elevated in patients with chronic kidney disease (CKD). TMAO might represent a biomarker that links the environment and microbiota with CKD. This Review summarizes data suggesting a link between the gut microbiota and derived metabolites with food intake patterns, metabolic alterations, and chronic CMDs.
Article
Full-text available
Assessment and characterization of gut microbiota has become a major research area in human disease, including type 2 diabetes, the most prevalent endocrine disease worldwide. To carry out analysis on gut microbial content in patients with type 2 diabetes, we developed a protocol for a metagenome-wide association study (MGWAS) and undertook a two-stage MGWAS based on deep shotgun sequencing of the gut microbial DNA from 345 Chinese individuals. We identified and validated approximately 60,000 type-2-diabetes-associated markers and established the concept of a metagenomic linkage group, enabling taxonomic species-level analyses. MGWAS analysis showed that patients with type 2 diabetes were characterized by a moderate degree of gut microbial dysbiosis, a decrease in the abundance of some universal butyrate-producing bacteria and an increase in various opportunistic pathogens, as well as an enrichment of other microbial functions conferring sulphate reduction and oxidative stress resistance. An analysis of 23 additional individuals demonstrated that these gut microbial markers might be useful for classifying type 2 diabetes.
Article
Full-text available
In this study, we compared the fecal microbiota and metabolomes of 26 healthy subjects before (HS) and after (HSB) 2 months of diet intervention based on the administration of durum wheat flour and whole-grain barley pasta containing the minimum recommended daily intake (3 g) of barley β-glucans. Metabolically active bacteria were analyzed through pyrosequencing of the 16S rRNA gene and community-level catabolic profiles. Pyrosequencing data showed that levels of Clostridiaceae (Clostridium orbiscindens and Clostridium sp.), Roseburia hominis, and Ruminococcus sp. increased, while levels of other Firmicutes and Fusobacteria decreased, from the HSB samples to the HS fecal samples. Community-level catabolic profiles were lower in HSB samples. Compared to the results for HS samples, cultivable lactobacilli increased in HSB fecal samples, while the numbers of Enterobacteriaceae, total coliforms, and Bacteroides, Porphyromonas, Prevotella, Pseudomonas, Alcaligenes, and Aeromonas bacteria decreased. Metabolome analyses were performed using an amino acid analyzer and gas chromatography-mass spectrometry solid-phase microextraction. A marked increase in short-chain fatty acids (SCFA), such as 2-methyl-propanoic, acetic, butyric, and propionic acids, was found in HSB samples with respect to the HS fecal samples. Durum wheat flour and whole-grain barley pasta containing 3% barley β-glucans appeared to be effective in modulating the composition and metabolic pathways of the intestinal microbiota, leading to an increased level of SCFA in the HSB samples.
Article
Diabetes and obesity are two metabolic diseases characterized by insulin resistance and a low-grade inflammation. Seeking an inflammatory factor causative of the onset of insulin resistance, obesity, and diabetes, we have identified bacterial lipopolysaccharide (LPS) as a triggering factor. We found that normal endotoxemia increased or decreased during the fed or fasted state, respectively, on a nutritional basis and that a 4-week high-fat diet chronically increased plasma LPS concentration two to three times, a threshold that we have defined as metabolic endotoxemia. Importantly , a high-fat diet increased the proportion of an LPS-containing microbiota in the gut. When metabolic endotoxemia was induced for 4 weeks in mice through continuous subcutaneous infusion of LPS, fasted glycemia and insulinemia and whole-body, liver, and adipose tissue weight gain were increased to a similar extent as in high-fat–fed mice. In addition, adipose tissue F4/80-positive cells and markers of inflammation, and liver triglyceride content, were increased. Furthermore, liver, but not whole-body, insulin resistance was detected in LPS-infused mice. CD14 mutant mice resisted most of the LPS and high-fat diet–induced features of metabolic diseases. This new finding demonstrates that metabolic endotoxemia dysregulates the inflammatory tone and triggers body weight gain and diabetes. We conclude that the LPS/CD14 system sets the tone of insulin sensitivity and the onset of diabetes and obesity. Lowering plasma LPS concentration could be a potent strategy for the control of metabolic diseases.
Article
Background and objectives: The generation of key uremic nephrovascular toxins, indoxyl sulfate (IS), and p-cresyl sulfate (PCS), is attributed to the dysbiotic gut microbiota in CKD. The aim of our study was to evaluate whether synbiotic (pre- and probiotic) therapy alters the gut microbiota and reduces serum concentrations of microbiome-generated uremic toxins, IS and PCS, in patients with CKD. Design, setting, participants, & measurements: Predialysis adult participants with CKD (eGFR=10-30 ml/min per 1.73 m(2)) were recruited between January 5, 2013 and November 12, 2013 to a randomized, double-blind, placebo-controlled, crossover trial of synbiotic therapy over 6 weeks (4-week washout). The primary outcome was serum IS. Secondary outcomes included serum PCS, stool microbiota profile, eGFR, proteinuria-albuminuria, urinary kidney injury molecule-1, serum inflammatory biomarkers (IL-1β, IL-6, IL-10, and TNF-α), serum oxidative stress biomarkers (F2-isoprostanes and glutathione peroxidase), serum LPS, patient-reported health, Gastrointestinal Symptom Score, and dietary intake. A prespecified subgroup analysis explored the effect of antibiotic use on treatment effect. Results: Of 37 individuals randomized (age =69±10 years old; 57% men; eGFR=24±8 ml/min per 1.73 m(2)), 31 completed the study. Synbiotic therapy did not significantly reduce serum IS (-2 μmol/L; 95% confidence interval [95% CI], -5 to 1 μmol/L) but did significantly reduce serum PCS (-14 μmol/L; 95% CI, -27 to -2 μmol/L). Decreases in both PCS and IS concentrations were more pronounced in patients who did not receive antibiotics during the study (n=21; serum PCS, -25 μmol/L; 95% CI, -38 to -12 μmol/L; serum IS, -5 μmol/L; 95% CI, -8 to -1 μmol/L). Synbiotics also altered the stool microbiome, particularly with enrichment of Bifidobacterium and depletion of Ruminococcaceae. Except for an increase in albuminuria of 38 mg/24 h (P=0.03) in the synbiotic arm, no changes were observed in the other secondary outcomes. Conclusion: In patients with CKD, synbiotics did not significantly reduce serum IS but did decrease serum PCS and favorably modified the stool microbiome. Large-scale clinical trials are justified.
Article
Also known as the "second human genome," the gut microbiome plays important roles in both the maintenance of health and the pathogenesis of disease. The symbiotic relationship between host and microbiome is disturbed due to the proliferation of dysbiotic bacteria in patients with chronic kidney disease (CKD). Fermentation of protein and amino acids by gut bacteria generates excess amounts of potentially toxic compounds such as ammonia, amines, thiols, phenols, and indoles, but the generation of short-chain fatty acids is reduced. Impaired intestinal barrier function in patients with CKD permits translocation of gut-derived uremic toxins into the systemic circulation, contributing to the progression of CKD, cardiovascular disease, insulin resistance, and protein-energy wasting. The field of microbiome research is still nascent, but is evolving rapidly. Establishing symbiosis to treat uremic syndrome is a novel concept, but if proved effective, it will have a significant impact on the management of patients with CKD.
Article
The gut microbiota plays an important role in human health by interacting with host diet, but there is substantial inter-individual variation in the response to diet. Here we compared the gut microbiota composition of healthy subjects who exhibited improved glucose metabolism following 3-day consumption of barley kernel-based bread (BKB) with those who responded least to this dietary intervention. The Prevotella/Bacteroides ratio was higher in responders than non-responders after BKB. Metagenomic analysis showed that the gut microbiota of responders was enriched in Prevotella copri and had increased potential to ferment complex polysaccharides after BKB. Finally, germ-free mice transplanted with microbiota from responder human donors exhibited improved glucose metabolism and increased abundance of Prevotella and liver glycogen content compared with germ-free mice that received non-responder microbiota. Our findings indicate that Prevotella plays a role in the BKB-induced improvement in glucose metabolism observed in certain individuals, potentially by promoting increased glycogen storage.