Next Article in Journal
Transition Mutations in the hTERT Promoter Are Unrelated to Potential i-motif Formation in the C-Rich Strand
Next Article in Special Issue
Synergistic Effect of Retinoic Acid and Lactoferrin in the Maintenance of Gut Homeostasis
Previous Article in Journal
Targeting Breast Cancer: The Familiar, the Emerging, and the Uncharted Territories
Previous Article in Special Issue
The Effects of a High-Fat Diet on Inflammatory Bowel Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Prebiotics Progress Shifts in the Intestinal Microbiome That Benefits Patients with Type 2 Diabetes Mellitus

1
Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
2
OzStar Therapeutics Pty Ltd., Pennant Hills, NSW 2120, Australia
3
Gold Coast University Hospital, Southport, QLD 4215, Australia
4
Prince of Wales Clinical School, University of New South Wales, Sydney, NSW 2052, Australia
*
Author to whom correspondence should be addressed.
Biomolecules 2023, 13(9), 1307; https://doi.org/10.3390/biom13091307
Submission received: 1 July 2023 / Revised: 22 August 2023 / Accepted: 23 August 2023 / Published: 25 August 2023
(This article belongs to the Special Issue Human Gut Microbiome and Diet in Health and Diseases)

Abstract

:
Hypoglycemic medications that could be co-administered with prebiotics and functional foods can potentially reduce the burden of metabolic diseases such as Type 2 Diabetes Mellitus (T2DM). The efficacy of drugs such as metformin and sulfonylureas can be enhanced by the activity of the intestinal microbiome elaborated metabolites. Functional foods such as prebiotics (e.g., oligofructose) and dietary fibers can treat a dysbiotic gut microbiome by enhancing the diversity of microbial niches in the gut. These beneficial shifts in intestinal microbiome profiles include an increased abundance of bacteria such as Faecalibacterium prauznitzii, Akkermancia muciniphila, Roseburia species, and Bifidobacterium species. An important net effect is an increase in the levels of luminal SCFAs (e.g., butyrate) that provide energy carbon sources for the intestinal microbiome in cross-feeding activities, with concomitant improvement in intestinal dysbiosis with attenuation of inflammatory sequalae and improved intestinal gut barrier integrity, which alleviates the morbidity of T2DM. Oligosaccharides administered adjunctively with pharmacotherapy to ameliorate T2DM represent current plausible treatment modalities.

1. Introduction

The intestinal microbiota presents a complex ecosystem consisting of bacteria, enteric viruses, archaea, fungi, and protozoa [1,2]. The structure of the intestinal microbiota is influenced by host genetics and environmental factors. Recent studies have shown that the gut microbiota exhibits a complex contribution to overall health, by actively regulating metabolic functions through genes, proteins, and metabolites. Furthermore, multiple factors contribute to the establishment of the human gut microbiota during infancy [3]. The main factors that are the predominant drivers in shaping the intestinal microbiome include diet across a lifetime; intestinal bacteria have a pivotal role in maintaining a metabolic and immune function equilibrium that protects against pathogenic insults.
Any subsequent alterations in the composition of the gut bacterial profile (dysbiosis) have been associated with the pathogenesis of numerous inflammatory diseases and infections [3]. Moreover, based on current evidence, it has been recently postulated and encouraged that novel strategies should integrate the administration of probiotics with conventional anti-cancer therapies [4].
The current scientific literature suggests that as the intestinal microbiota composition is observed to differ between individuals and is contingent on a variety of factors such as genetics and diet, a percentage of individuals may harbour intestinal bacteria that have been reported to be associated with pro-inflammatory effects, while others may harbour intestinal bacteria with anti-inflammatory effects [5]. Biochemical techniques that allow for the enhanced characterization of the intestinal microbiome have provided further evidence of the pro-inflammatory and anti-inflammatory nature of the gut cohort of bacteria in health and disease [5]. The continuous improvement in knowledge relevant to the inflammatory pathways that interact with bacteria will further elucidate factors that underpin the varying presentations of the same disease and the varied responses reported to the same treatment in different individuals [5]. What becomes biologically and clinically plausible is the administration of anti-inflammatory microbes that can be formulated in probiotic formulations and used in therapies with or without prebiotics [5].
The administration of oral probiotic bacteria has been demonstrated to interact with numerous intestinal structures, including intestinal epithelial cells and immune cells associated with the lamina propria, through Toll-like receptors that then induce the production of a plethora of cytokines/chemokines [6,7,8]. Macrophage chemoattractant protein 1, produced by intestinal epithelial cells, produces signals that are interpreted by other immune cells, leading to the activation of the mucosal immune system. Such activation has been reported to be characterized by increased immunoglobulin A+ cells of the intestine, bronchus, and mammary glands and the activation of T cells [6,8].
Furthermore, probiotics have been reported to activate regulatory T cells that release an anti-inflammatory interleukin (i.e., IL-10). What has been reported is that probiotics have multiple effects that reinforce the intestinal epithelial barrier integrity, coupled with increases in mucins, tight junction proteins, and Goblet and Paneth cells [6,7]. Additionally, probiotics have been proposed to modulate the intestinal microbiota by maintaining a healthy pro-inflammatory to anti-inflammatory balance that continuously suppresses the overgrowth of potential pathogenic bacteria in the intestines. Alternatively, it has been demonstrated that the long-term administration of probiotics does not affect the intestinal homeostasis. The viability of probiotics is crucial in their interactions with intestinal epithelial cells and macrophages, which are skewed toward a favorable innate immune response. The important role of macrophages in the immune response progresses without inducing an inflammatory pattern, with minor increases in the cellularity of the lamina propria [6,7].
Of interest is the role that probiotics may have in activating the microbicidal actions of peritoneal and spleen macrophages to protect against the action of different pathogens [6]. Furthermore, it has been also reported that in malnutrition models, such as undernourishment and obesity, probiotic formulations have been able to increase both intestinal and systemic immune responses. Additionally, probiotic bacteria may also contribute to recovering the histology of both the intestine and the thymus that have been damaged due to poor nutrition choices. The administration of probiotic bacteria has a long history, and the emerging evidence shows that it is a safe and natural strategy for allergy prevention and treatments [6]. Moreover, different mechanisms such as those proposed to generate cytokines from activated pro-T-helper type 1 cells have been reported to favor the production of IgG instead of IgE [6].
A recent review reports that intestinal microbiota dysbiosis has been shown to have adverse health effects that will lead to a variety of chronic disease progressions [9]. The subsequent regulation and mechanism of involvement of the intestinal microbiota have been reported in neurodegenerative diseases, cardiovascular diseases, metabolic diseases, and gastrointestinal diseases [9]. Furthermore, a recent review has reported that experimental models of inflammatory bowel disease have strongly suggested that while intestinal bacteria often progress immune activation, chronic inflammatory responses in turn can shape the intestinal microbiota and contribute to dysbiosis in inflammatory bowel diseases [10].
It is generally accepted that in genetically susceptible individuals, inflammatory bowel diseases can result from the dysregulation of immune responses to environmental and/or microbial agents [11]. Of interest is a recent laboratory murine study that administered the antibiotics rifaximin (i.e., a dose of 50 mg/kg/dose) and/or Mutaflor (109 CFU/dose) given intragastrically once a day to investigate the healing effect of acetic acid-induced colitis [12]. The study reported that the antibiotic significantly accelerated the healing of colonic damage. Moreover, the administration of the antibiotic significantly reduced the concentration of the proinflammatory cytokine TNF-α, as well as the activity of myeloperoxidase in the colonic mucosa of rats [12]. Mutaflor (i.e., a probiotic formulation) given alone had no significant effect on the activity of colitis. However, when Mutaflor was administered in combination with the antibiotic, there was a significantly enhanced therapeutic effect of the antibiotic. The authors concluded that the antibiotic and probiotic formulations exhibited synergic anti-inflammatory and therapeutic effects in acetic-acid-induced colitis in rats [12].
A growing body of evidence reports that intestinal bacteria can influence a plethora of hormones [13]. For example, decreased abundance in microbial diversity has been linked with high thyroid stimulating hormone (TSH) levels [14]. Furthermore, an increase in TSH concentrations may cause the production of lower levels of thyroid hormones T3 and T4, which can at times progress to hypothyroidism. An imbalanced intestinal microbiome has also been associated with hypothyroidism [15]. Studies document that common symptoms of hypothyroidism include weight gain, sensitivity to cold, dry skin, constipation, and poor memory [16]. The hormone estrogen is also subject to bacterial actions. Studies report that the intestinal microbiota is a key regulator of the level of circulating estrogen in the systemic circulation [17]. In addition, the intestinal microbes can produce beta-glucuronidase, an enzyme that can convert estrogen into its active forms [17]; it was also noted that intestinal dysbiosis can alter the amount of active estrogen in the circulation. Furthermore, research reports that there exists a specific group of bacteria termed the estrobolome [18]. The estrobolome has been reported to consists of bacterial genes that are capable of metabolizing estrogens, an important factor given that estrogen is a potent promoter of tissue growth throughout the body [18].
Another important metabolite that is associated with gut bacteria is serotonin [19]. Serotonin is a precursor for the formation of melatonin, a hormone that has been reported to regulate mood [19]. Of significant clinical interest is that studies have reported that shift workers tend to present with gut dysbiosis. Hence, insufficient sleep can cause negative effects on the intestinal microbiome [19]. Further, shift work has been shown to increase inflammatory responses and the risk of obesity, metabolic syndrome, and T2DM.
Additional hormones such as cortisol, epinephrine, and norepinephrine are hormones that are associated with an alert response [20]. These hormones are released during the flight-or-flight response, leading to heart rate and blood pressure elevations, which can subsequently increase hormone levels. However, when these hormone levels remain high for prolonged periods, the dynamics of the intestinal microbiome can be altered [20]. High levels of stress hormones have also been shown to trigger adverse gene expression in gut microbes [20]. In animal studies, high epinephrine and norepinephrine levels can stimulate infections by increasing the virulence of some pathogens [20].
A chronic increase in intestinal permeability that has been termed leaky gut has been observed in the patients and animal models of metabolic diseases (e.g., T2DM) [21]. This state often correlates with a metabolic disease state [22]. Recent reports have documented that the intestinal microbiota affects intestinal and systemic health via bacterial-generated metabolites, especially short-chain fatty acids and lipopolysaccharides, which can trigger and maintain a leaky gut [22].
The endotoxin lipopolysaccharide (LPS) is a component of Gram-negative bacteria and is responsible for sepsis and neonatal mortality [23], yet in experimental studies, low levels of LPS show tissue protection. In a recent murine study, the effects of LPS, which was applied to suckling rats on the pancreas of adult animals, were investigated [23]. The study concluded that the endotoxemia that was induced in suckling rats by the intraperitoneal application of LPS from Escherichia coli or Salmonella typhi in the early period of life with LPS reduced histological manifestations of acute pancreatitis. Moreover, pancreatic weight and plasma lipase activity were decreased, and SOD concentration was reversed and accompanied by a significant reduction in lipid peroxidation products in the pancreatic tissue [23]. In the pancreatic acini, there were observed significant increases in protein signals for toll-like receptor 4 and for heat shock protein 60. In addition, the signal for the CCK1 receptor was reduced and pancreatic secretory responses to caerulein were decreased, whereas basal enzyme secretion was unaffected [23]. Hence exposing suckling rats to endotoxin had an impact on the pancreas in the adult organism.
T2DM is a group of chronic endocrine and metabolic disorders characterized by defects in insulin production, secretion, and signaling that are insufficient to maintain a balanced blood glucose level. The interaction between the gut microbiome, dietary practices, and the activity of mucosal immunity influences the progression of T2DM. A significant body of research evidence has documented that the intestinal microbiome is associated with metabolic disease and that the progression of intestinal dysbiosis leads to gut metabolic dysregulation [24,25].
Studies have shown that functional foods administered as prebiotics and/or probiotics have led to beneficial alterations of the intestinal microbiome in T2DM, hence improving glycemic control [26,27]. In addition, recent studies explored the co-administration of probiotics and prebiotics as adjuncts to pharmacotherapy, also providing an effective treatment strategy in managing hyperglycemia symptoms in T2DM [27,28].
Functional foods from plants have a long history of promoting health benefits, primarily due to the supply of compounds that, at least in part, can offer protective effects, as exemplified by fruit and vegetable-rich diets [29]. The study of functional foods in human nutrition has become a fundamental issue in the prevention and management of metabolic disorders such as T2DM.

2. Pathogenesis of Diabetes Mellitus: Link between the Intestinal Microbiome Dysbiosis and Insulin Resistance

The exact factors that drive the development of metabolic diseases remain elusive, as most metabolic diseases can be associated with both a Western-type diet and adverse microbiome modifications such as dysbiotic gut. These intestinal microbiome shifts are characterized by reduced diversity, enrichment of opportunistic pathogens, and an imbalanced ratio of beneficial to detrimental metabolites [30]. Various distinct pathophysiologic abnormalities have been associated with T2DM. A decreased peripheral glucose uptake (i.e., mainly from skeletal muscle) in combination with an augmented endogenous glucose production are the characteristic hallmark features of insulin resistance [31,32]. Furthermore, increased lipolysis, elevated systemic free fatty acid levels, and the accumulation of intermediary lipid metabolites strongly contribute to further increases in glucose output, reducing peripheral glucose utilization, and impairment of beta-cell function [32,33]. A recent review summarized data from epidemiological studies that noted that patients diagnosed with T2DM have a two-fold increased risk for non-alcoholic fatty liver disease (NAFLD) development and vice versa [31]. The presence of NAFLD] is now considered an integral part of the insulin resistant state [31]. The traditional concepts of glucotoxicity and lipotoxicity, which encompass the process of beta cell deterioration in response to chronic elevations in glucose and lipids, also include all nutrients, giving rise to the term nutri-toxicity [32]. The delayed transport of insulin across the microvascular system is also partially responsible for the development of tissue insulin resistance.
Furthermore, Solis-Herrera and colleagues [32] report that compensatory insulin secretion by pancreatic beta cells could initially maintain normal plasma glucose levels. Yet they also report that beta cell function is already compromised and abnormal during the early stages of T2DM and progressively becomes aggravated over time.
Solis-Herrera and colleagues have suggested that during the post-prandial period, there is an inappropriate release of glucagon from the alpha cells of the pancreas [32]. Consequently, Solis-Herrera et al. posit that this is probably due to impaired insulin secretion as well as an excessive secretion of glucagon in patients diagnosed with T2DM. These biochemical effects would be secondary events to an incretin defect, where the latter is defined as a primary inadequate release or response to the gastrointestinal incretin hormones upon meal ingestion. Therefore, it is interesting to note that the intestinal microbiome may be complicit in the hormonal and metabolic disturbances that ensue and that are observed in T2DM.
Glucose toxicity research refers to higher than normal concentrations of glucose in cells and tissues that have been associated with adverse health effects [32,34]. The most common effect reported is that glucose toxicity presents as the ability of excess circulating systemic glucose to impair insulin secretion and action. A resultant aggravated cycle of declined glycemic control in T2DM ensues [32,34]. Glucose toxicity has been expanded to encompass other effects of hyperglycemia such as the complications of diabetes [32]. Furthermore, it has been suggested that such terminology (i.e., glucose toxicity) be applied to those adverse effects of high glucose concentrations that are specific for glucose or its metabolites (i.e., are not reproduced by hyperosmolality) [34]. McClain has also noted that the term toxicity may be misleading as some of the ultimately harmful effects of excess systemic glucose levels may represent normal physiologic responses rather than a significant deleterious biochemical effect on cells by non-physiologic mechanisms [34].
Moreover, investigations relevant to the intestinal organoids have been used to analyze the differentiation of enteroendocrine cells and to manipulate their density for treating T2DM [35]. Such molecular mechanisms may have direct clinical implications. In addition, Filipello and colleagues [35] recently showed that glucotoxicity can impair L-cell differentiation, an important factor associated with decreased intestinal stem cell proliferative capacity [35]. We note that this study provided the identification of new targets involved in new molecular signaling mechanisms that may be impaired by glucotoxicity and that these targets could be a useful tool for the treatment of T2DM [35]. Obesity is a risk factor for various metabolic disorders such as T2DM and cardiovascular disease [36]. The two dominant phyla in the intestines comprise the Firmicutes and Bacteriodetes [37]. Reports show an existing link between the intestinal microbiome and obesity, with an attendant increase in the abundance of Firmicutes and a decrease in the diversity of Bacteroidetes [37,38]. Obesity and the complicit intestinal microbiome in T2DM links host molecules that induce intestinal microbiota dysbiosis and immune and pro-inflammatory responses, with intestinal microbial metabolites being involved in the pathogenesis of the disease [39]. The T2DM metabolic syndrome of abnormal lipid and glucose metabolism is associated with numerous dysfunctional physiological complications [40]. T2DM has been characterized by pancreatic beta-cell dysfunction and peripheral insulin resistance, leading to defects in glucose metabolism [41]. In addition, a dysbiotic intestinal microbiome has been reported to be complicit in the pathogenesis of T2DM by maintaining the gut in a chronic low-grade inflammatory state [39,42]. The earliest proposed strategy to manipulate the intestinal microbiome for a health benefit was the use of probiotics [43]. A recent review has directed new perspectives in the treatment of T2DM, with the goal being to achieve an effective adjustment of blood glucose and blood lipid levels as well as body weight in patients diagnosed with T2DM to healthy profiles [44]. The administration of synbiotic formulations (i.e., prebiotics + probiotics) can also improve glucose and lipid profiles in patients diagnosed with T2DM [44]. Studies with prebiotics (without probiotics) have also shown efficacy in patients diagnosed with T2DM (Table 1).
Could prebiotics that significantly influence the gut microbiome, administered as adjuvants to pharmacotherapy, maintain favorable indexes of lipid profile, blood pressure, and fasting blood glucose in patients with T2DM? The application of adjuvant prebiotics with oral hypoglycemic medications could be a new management strategy for lipid and glucose profiles and blood pressure management in T2DM. Two studies with prebiotics and sulfonylureas or metformin have reported improvements in the associated clinical parameters (Table 2). This has become a plausible research initiative given that metformin has been shown in a number of studies with obese and/or T2DM-diagnosed patients to progress gut microbiome shifts favorable to improved glycemic control [45]. Specifically, it was reported that the overall results showed that metformin significantly shifted the proportion of the phyla Bacteroidetes and Verrucomicrobia and the genera Akkermansia, Bacteroides, and Escherichia. Shifts in the intestinal microbiome remain consistent with the view that gut bacteria influence the hypoglycemic effect of metformin. Mechanistically through the improved integrity of the intestinal epithelial barrier, the production of SCFAs (e.g., butyrate), the regulation of bile acid metabolism, and improved glucose homeostasis [45].
Oligosaccharides are a type of carbohydrate naturally found in many types of plant foods. Oligosaccharides are low-level polysaccharides formed by the linkage of 2–10 monosaccharides through glycosidic bonds. Common oligosaccharides include chitosan oligosaccharides, xylo-oligosaccharides, konjac glucomannan, and brown algae oligosaccharides [46]. In a clinical trial with women diagnosed with T2DM, an oligofructose was reported to improve inflammatory markers [47]. In a systematic review, it was posited that oligosaccharides could offer benefit as a gut microbiome therapeutic strategy for T2DM [48]. In view of this, a recent systematic review and meta-analysis [49] concluded that oligosaccharides can provide beneficial effects for the markers of glycemic control, specifically fasting blood glucose, fasting blood insulin, glycated hemoglobin, homeostasis model assessment of insulin resistance, and quantitative insulin sensitivity index.

Fiber, Prebiotics, and Intestinal Bacteria

There has been a long appreciation of dietary fiber as being beneficial for intestinal health and improving symptoms of gut progressed inflammatory sequelae [50]. Furthermore, dietary fiber is reported to be a group of heterogenous compounds that are neither digested nor absorbed in the intestines [51]. Categories of fiber, including inulin, fructo-oligosaccharides, and other oligosaccharides, have been included in food labels. Consequently, oligosaccharides have been considered to be the best-known example of a prebiotic, being a selectively fermented ingredient that allows specific changes, both in the composition and/or activity of the intestinal microbiome, which may confer health benefits on the host [50].
A recent review tabulates resistant oligosaccharides (e.g., fructans (fructooligosaccharides, oligofructose (OF), and inulin) and galactans) as well documented prebiotics [51]. Additional fibers have been posited as candidates of prebiotics or as having the potential to be classified as prebiotics, while other fibers have no prebiotic effect [51]. Furthermore, studies that have provided evidence for inulin, OF, lactulose, and resistant starch (RS) meet the definition of fibers that encourage bacteria from the Bifidobacterium genus. Additionally, isolated carbohydrates and carbohydrate-containing foods, including galactooligosaccharides (GOS), transgalactooligosaccharides (TOS), polydextrose, wheat dextrin, acacia gum, psyllium, banana, whole grain wheat, and whole grain corn, have also been documented as providing a prebiotic effect in the gut [50].
Prebiotics are defined as a group of compounds that have been associated with gut health and that are mediated, at least in part, through degradation products by microorganisms [52]. The chemical structure of prebiotics and the bacterial composition of the intestines determine the fermentation products produced [53,54]. Dietary fiber is found mainly in fruits, vegetables, whole grains, and legumes, and their effects on health mimic those of prebiotics on human health [54]. Numerous compounds have been investigated to determine if, as prebiotics, these functional non-digestible foods can influence the diversity of the intestinal microbiome to promote metabolic health in the gut [53,55] (Table 1). The most common forms of prebiotics that have been studied include fructo-oligosaccharides, galacto-oligosaccharides, and trans-galacto-oligosaccharides [56,57]. These prebiotics are not digested by the host enzymes/tissues. When taken orally, prebiotics travel to the intestines, where they are then metabolized by microorganisms to be used as energy sources [58].
The biochemical process of the fermentation of fiber-rich compounds by the intestinal microbiome yields short-chain fatty acids (SCFAs), including acetate, lactate, butyrate, and propionate [59,60,61]. The net effects of the increased production of SCFAs in the intestines are the lowering of the luminal pH [62], improved availability of calcium and magnesium, and inhibition of potentially pathogenic bacteria [63]. Both Bifidobacterium and Lactobacillus produce acetate (and lactate), and, as such, contribute to the SCFA-mediated health benefits that prebiotics have been reported to provide. Notwithstanding, members of these genera of microorganisms do not produce butyrate and/or propionate [64]. Moreover, the important function of the bacterial species from the Bifidobacterium genus is that these microbes contribute to gut homeostasis and host health. This is achieved through the production of acetate and lactate during carbohydrate fermentation, which are organic acids that in turn can be converted into butyrate by other colon bacteria through cross-feeding interactions [65].
A recent study has investigated how SCFAs modulate the intestinal microbiome and progress their functionality [66]. The results provided important information on how prebiotics elevate the colonic level of specific SCFAs for therapeutic benefit. Various human studies with healthy children and adults have demonstrated the effect that prebiotics have on the abundance and dynamics of microbial species in the intestines (Table 1). Clinical studies that have been conducted have also reported significant favorable shifts in the intestinal microbiome. In most of the studies, shifts towards increases in the abundance of Bifidobacterium and Lactobacillus genera have been reported.
Table 1. Prebiotics shift the intestinal microbiome: human studies.
Table 1. Prebiotics shift the intestinal microbiome: human studies.
PREBIOTICS
(n)
REFERENCE
TREATMENT TIMEDOSE
ADMINISTERED
FLUIDFAECAL INTESTINAL MICROBIAL SHIFTS BY
PREBIOTICS VERSUS CONTROLS
STUDIES WITH CHILDREN
PDX/FOS *
[n = 77]
[67]
2-weeks4.17 g PDX|
0.45 g FOS
O.D.
in waterBifidobacterium and Lactobacillus
AX
[n = 10]
[68]
48 h10 g/L fluidin waterLactobacillus
STUDIES WITH ADULTS
Inulin
[n = 17 W]
[69]
8 days20 g/day to 40 g/dayin waterBifidobacterium
Enterococci|Enterobacteriaceae
Inulin
[n = 10]
[70]
2-weeks8 g/day
B.I.D.
in waterBifidobacterium|↑ Clostridia
GOS
[n = 18]
[71]
3-weeks2.5 g/day
5 g/day
10 g/day
edible chews↑ abundance Bifidobacterium
Faecalibacterium prausnitzii
Bacteroides
PDX
[n = 20 M]
[72]
3-weeks21 g/day
O.D.
mixed foodFaecalibacterium | Phascolarctobacterium|
Dialister
PDX
[n = 15]
[73]
3-weeks8 g/day
O.D.
powderRuminococcus intestinalis|
Clostridium clusters I, II and IV,
↓ abundance Lactobacillus|
Enterococcus group
Lactulose
[n = 12]
[74]
4-weeks20 g/day
B.I.D.
mixed foodBifidobacterium|Lactobacillus
Synthetic
2′-O-fucosyllactose and/or lacto-N-neotetraose
[n = 44]
[75]
2-weeks5 10 20 g/dayDietary barsActinobacteria | Bifidobacterium
Firmicutes|Proteobacteria
Wheat Bran or Maltodextrin
[n = 20]
[76]
3-weeks
Cross-over
study
10 g/dayDietary barsBifidobacterium adolescentis
PDX + SCF or
placebo
[n = 21]
[77]
3-weeks
Cross-over
study
21 g/day PDX
21 g/day SCF
Dietary barsWith increased total corn fiber consumed from 10 g to 50 g/day
Optimum mean daily consumption of 30–35 g/day
Shift of the Bacteroidetes:Firmicutes ratio positively correlated to total dietary fiber intake.
Soluble Corn Fiber
[n = 28 F]
[78]
4-weeks/dose
[12-weeks]
0 10 20 g/dayin waterClostridium
↑ unclassified Clostridiaceae
XOS
[n = 12 W
[n = 11 M]]
[79]
8-weeks1.4 g/day or
2.8 g/day
O.D.
capsuleBacteroides fragilis|Bifidobacterium
XOS
[n = 13]
[80]
3-weeks4 g/daymixed foodBifidobacterium species
Inulin or Maltodextrin
[n = 44]
[81]
8-weeks
Cross-over study
12 g/dayin water↑ relative abundances
Anaerostipes|Bilophila|Bifidobacterium
Crystalline Maize
[Treat Arm 1}
[n = 5 W 5 M]
4-weeks dose escalation0 10 20 35 50 g/day
10 g/d to 50 g/d
powder sachets in waterIndividualized effects with
B. adolescentis|Parabacteroides spp.|
Eisenbergiella spp.
(Maize)
Tapioca RS4
[n = 5 W 5M]
[Treat Arm 2]
Digestible Corn Starch
[n = 5 W 5 M]
[Placebo Arm]
Potato RS4
[n = 5 W 5 M]
[82]
[Treat Arm 3]
4-weeks dose escalation





4-weeks dose escalation
0 10 20 35 50 g/day
10 g/d to 50 g/d





0 10 20 35 50 g/day
10 g/d to 50 g/d
powder sachets in water



powder sachets in water
E. rectale (Maize)↑ P. distasonis|Clostridiales
(Tapioca extracted from cassava root)





E. rectale
P. distasonis
* O.D. = once per day; PDX/FOS = polydextrose/fructooligosaccharide; B.I.D. = twice per day; F = females; M = males; XOS = xylooligosaccharide; SCO = soluble corn fiber; RS4 = type-IV resistant starches; ↑ = increased; ↓ = decreased; → = no effect.
Other prebiotic compounds from dietary fiber can also encourage the production of butyrate and/or propionate in the intestines [83], which has been studied for its effects on human health. Consequently, prebiotics have been shown to increase significantly the colonic luminal concentration of SCFAs such as butyrate [54]. Butyrate is the primary energy source of differentiated intestinal epithelial cells (Figure 1), as its metabolism serves to consume oxygen, permitting the preservation of an anaerobic gut luminal environment that maintains the fitness of the strict anaerobes populating that part of the intestines [84].
In order to maintain homeostasis, the colonic epithelia are skewed toward oxidative phosphorylation, which results in high epithelial oxygen consumption [84]. Consequently, the resultant intestinal colonic epithelial hypoxia supports the maintenance of a local microbial community that is obligate anaerobic bacteria dominant, providing a beneficial conversion of fiber into fermentation products that can be absorbed by the host [84]. Alternatively, those conditions that alter the metabolism of the colonic epithelium, which increases epithelial oxygenation that drives an expansion of facultative anaerobic bacteria, can lead to gut dysbiosis in the colon. Enteric pathogens can then subvert colonocyte metabolism to escape the niche protection conferred by the gut microbiota [84].
Butyrate has also been reported to act as a histone deacetylase (HDAC) inhibitor, exhibiting pleiotropic modifications in biochemical transcription and host physiology [87] and exerting an influence on the activation of anti-inflammatory Treg cells [84].
Consequently, SCFAs have been reported to have multiple effects on the host. For example, reports have documented that butyrate can stimulate the synthesis of host defense peptides via intestinal epithelial cells [88]. The mechanism is via the interactions of butyrate with G protein-coupled receptor 43 (GPR43). GPR43 activates the Jun N-terminal kinase, mitogen-activated protein kinase, extracellular signal-regulated kinase pathways, and cell growth pathways [89]. Butyrate can also influence intestinal epithelial development and barrier integrity [59]. Given that SCFAs can diffuse into the systemic circulation through gut enterocytes, this translocation allows metabolites generated from the bacterial fermentation of prebiotics to affect not only the intestinal physiology but also that of distant site organs (e.g., the liver) [18].
Other SCFAs such as propionate can affect a subset of CD4+ T helper cells: the T helper 2 (TH2) cells [90]. TH2 cells facilitate the activation and maintenance of antibody-mediated immune responses against extracellular parasites, bacteria, allergens, and toxins [90]. TH2 cells mediate these functions by co-operating and/or producing various cytokines such as IL-4, IL-5, IL-6, IL-9, IL-13, and IL-17E (IL-25) [90]. This, in turn, may stimulate macrophages in the respiratory tract as well as dendritic cells in the bone marrow [91]. Peptidoglycan (i.e., the basic structure in nearly all bacterial cell walls) [92] may be considered as an endogenous gut prebiotic product that can stimulate the innate immune system against pathogenic microorganisms [93]. In gut bacteria that inhabit the intestines of humans, when considering the bacterial cell wall component peptidoglycan, it is viewed as a key bacterial cell component. In immunology, peptidoglycans are conserved pathogen-associated molecular patterns (PAMP) for which the innate immune system has evolved sensing mechanisms [93]. Further, peptidoglycans have been reported to be a direct target for innate immune responses and for regulating the accessibility of other PAMP for additional innate immune responses.

3. Functional Foods for T2DM

From birth there is a complex and dynamic microbial assault on all mucosal surfaces of the host to achieve colonization with an obligatory progression to immunological tolerance and metabolic homeostasis [94]. These events can be enhanced with the administration of functional foods such as prebiotics and probiotics that have been reported to induce immunomodulatory and metabolic balancing activities in the intestines [86,95]. Different types of prebiotics can influence the intestinal microbiome by stimulating the growth of various indigenous bacterial communities. The combined research evidence from laboratory, animal, and human studies has shown that when prebiotics have been administered, there has been a subsequent gut enrichment of commensal species that promote health, such as those from the genera Lactobacillus and Bifidobacterium [96,97].
Oligosaccharides have been reported to improve inflammatory responses in the gut [98]. The intestinal microbiome has been reported to be significantly associated with gross adverse shifts in the structure of the intestinal microbiome. Furthermore, reports showed that the genera Ruminococcus, Fusobacterium, and Blautia were positively associated with T2DM, whereas the genera Bifidobacterium, Bacteroides, Faecalibacterium, Akkermansia, and Roseburia were negatively associated with T2DM [25]. Studies have shown that oligosaccharides in the intestine can be fermented and utilized by various bacteria, including Streptococcus [99], Escherichia coli [100], and Clostridium species [101]. A small clinical study explored the adjunctive administration of oligofructose with sulfonylurea [28], which increased the abundance of Bifidobacterium, a genus of intestinal bacteria, and reported the improvement of glycemic control in T2DM [102]. Bifidobacterium are reported to be acetate and propionate producers of SCFAs [103]. As such, intestinal bacteria from the genera Faecalibacterium, Akkermansia, and Roseburia utilize these SCFAs as energy sources while secreting butyrate, subsequently improving insulin resistance and glucose metabolism.
The adjunctive treatment for 6 months with oligofructose prebiotic for patients diagnosed with T2DM while prescribed sulfonylurea monotherapy resulted in clinically significant improvements in fasting blood glucose, postprandial blood glucose, area under the curve for serum glucose, and HbA1c. These improvements coincided with an increase in the endogenous insulin and c-peptide levels, collectively leading to significant improvements in pancreatic beta cell function. These improvements in glycemic control were associated with changes in the gut microbiome dynamics and an increase in beneficial and a reduction in detrimental bacteria that have previously been found to be associated with T2DM (Table 2) [28].
Table 2. Adjuvant prebiotics with and without pharmacotherapy for the management of T2DM. human studies.
Table 2. Adjuvant prebiotics with and without pharmacotherapy for the management of T2DM. human studies.
PREBIOTICS
(n)
REFERENCE
TREATMENT TIMEDOSE
ADMINISTERED
OUTCOMES
STUDIES with Prebiotics and T2DM
FOS *
[n = 27 W]
[47]
8-weeks10 g/day
O.D.
↓ Fasting plasma glucose
↓ Glycosylated hemoglobin
↓ Interleukin-6
↓ Tumor Necrosis Factor α
↓ Plasma Lipopolysaccharide
RS
[n = 17]
[104]
12-weeks40 g/day
O.D.
↓ Postprandial Glucose (meal tolerance test)
↑ Glucagon-Like Peptide-1
↓ Tumor Necrosis Factor
AX
[n = 15]
[105]
5-weeks49.2 g/day↓ Fasting Serum Glucose Level.
↓ Serum Glucose
↓ Insulin Level (2 h after oral glucose intake)
STUDIES with Prebiotics + Pharmacotherapy and T2DM
Prebiotic Fiber +
Sulfonylurea
[n= 30]
[28]
24-weeksSulfonylurea
+
OZ101 an OLF
13.5 or 27 g/d
T.I.D.
Low dose of 13.5 g/T.I.D. associated…
improved beta-cell function stabilization of glycemic control over 24 weeks.
Prebiotic Fiber +
Metformin
[n = 9 with 6 eligible]
[106]
1-week cross-over
4-week follow-up
Metformin (850 mg)
O.D. increased B.I.D.
+
Prebiotic Fiber
(BiomeBliss)
O.D.
Modest shifts in microbial composition
Proof-of-Concept feasibility for adjunctive metformin therapy
* O.D. = once per day; B.I.D. = twice per day; T.I.D. = three times per day; FOS = fructo-oligosaccharides; RS = resistant Starch; AX = arabinoxylan; OLF = oligofructose; ↑ = increased; ↓ = decreased.

4. Modulation of the Intestinal Microbiome in T2DM

The disruption of the epigenome poise may be causal for several pathologies and is a contributing factor to obesity and T2DM [107]. It is known that the epigenome includes DNA methylation, histone modifications, and RNA-mediated processes, where these mechanisms control gene activity and physiological development [108]. This can be exemplified through the adipose tissue, skeletal muscle, pancreatic islets, liver, and blood relations to obesity and T2DM [108,109].
In a sequence of important studies with T2DM patients and intestinal microbiota, researchers have reported the characterization of the intestinal microbiota with this metabolic disease (Table 3). Moderate gut microbial dysbiosis was applied to characterize the gut microbiota. A functional analysis has shown that glucose membrane transport, methane metabolism, heterogeneous biomass degradation, branched-chain amino acid transport and metabolism, and sulphate reduction pathways were enriched in patients with T2DM.
Table 3. The complex and progressive characterization of the intestinal microbiome associated with T2DM.
Table 3. The complex and progressive characterization of the intestinal microbiome associated with T2DM.
Patients with T2DM *
(n)
[Reference]
Faecal Microbiome
Analysis
Gut Microbiome Characterizations with T2DM
(n = 18/36 M|T2DM)
[110]
real-time quantitative PCR (qPCR)Plasma glucose association with
↓ abundance Firmicutes phylum
↓ abundance Clostridia class
↑ abundance Betaproteobacteria class
subgroup (n = 20)
tag-encoded amplicon
pyrosequencing of the V4
region of the 16S rRNA gene
+ve association with ratios of
Bacteroidetes > Firmicutes
Bacteroides-Prevotella > C. coccoides-E. rectale
(n = 345 and 23 T2DM)
[111]
MGWAS↓ abundance of butyrate-producing bacteria
Clostridiales sp. SS3/4|F. prausnitzii|
Roseburia intestinalis|Eubacteriumrectale|
Roseburia inulinivorans
T2DM characterized by a moderate degree of intestinal microbial dysbiosis
↑ abundance opportunistic pathogen bacteria
Bacteroides caccae|Clostridium hathewayi|
Clostridium symbiosum|Eggerthella lenta|
 Clostridium ramosum|Escherichia coli
↑ mucin-degrading bacteria Akkermansia muciniphila
↑ sulfate-reducing bacteria Desulfovibrio sp. 3_1_syn3
(n = 2595 W|
9.5% T2DM
14.4% IGT
[112]
SgSeq faecal meta-genome
sample n = 145 W
normal|impaired|
diabetic glucose control
Further confirmation
functional alterations of the gut microbiome
reflecting changes in the intestinal environment of T2DM
Patients with T2DM
↑ abundance 4 Lactobacillus spp. L. gasseri highest
↓ abundance 5 Clostridium spp. Roseburia|
 Faecalibacterium prausnitzii|
highly discriminant for T2DM
butyrate producers
linked with improved insulin sensitivity
* T2DM = type 2 diabetes mellitus; SgSeq = shotgun sequencing; MGWAS = metagenome-wide association study; M = men; W = women; ↑ = increased; ↓ = decreased.
T2DM and obesity are strongly linked with adiposopathy, along with adipose tissue dysfunction, which has been reported as a major factor in the etiology of metabolic dysfunction conditions [113]. Metabolic dysfunction promotes dysregulated glucose homeostasis and impaired adipogenesis, leading to the accumulation of ectopic fat, insulin resistance, and chronic inflammation [113]. The intestinal inflammatory cascade that is progressed with T2DM disrupts intestinal barrier homeostasis, increasing gut permeability, with a consequent increased risk of endotoxemia with the translocation of endotoxins, such as lipopolysaccharides (i.e., LPS), from Gram-negative bacteria (e.g., Clostridium perfringens) [114]. This significantly affects glucose metabolism and insulin resistance [114].
A recent systematic review analysed studies that had assessed the serum concentrations of the endotoxin lipopolysaccharide (LPS) or that of the lipopolysaccharide-binding protein (LBP) in diabetic patients and compared them to the serum of healthy individuals [115]. The review presented significant variability in the estimates that the studies reported of metabolic endotoxemia [115]. Most studies reported higher LPS or LBP concentrations in diabetic patients than in those of the healthy controls. Patients with type 1 diabetes mellitus (T1DM) and T2DM presented with higher mean fasting levels of LPS of 235.7% and 66.4%, respectively, as compared with non-diabetic patients. Advanced complications (e.g., such as macroalbuminuria) and disease onset were reported to exacerbate endotoxemia. Furthermore, antidiabetic medications interestingly decreased fasting LPS concentrations [115]. Among the medications, rosiglitazone and insulin presented with higher and lower effects, respectively, when compared with other pharmaceutical treatments. T1DM and T2DM appeared to increase metabolic endotoxemia [115]. The authors concluded that numerous confounders (i.e., diet, age, medication, smoking, and obesity) can influence both diabetes and endotoxemia expression [115].
An early review [116] reported that when LPS is present in the intestinal lumen, it can translocate the intestinal mucosal barrier and subsequently into the systemic circulation, causing metabolic endotoxemia [116]. LPS has the capacity to bind to toll-like receptor 4 (i.e., a receptor that belongs to the family of pattern recognition receptors (PRRs)).
There is subsequent activation of pro-inflammatory activity that can alter several stages of insulin signaling. Furthermore, the review reports that chronic exposure to the LPS endotoxin may contribute to weight gain and the progression of T2DM [117].
Obese and diabetic individuals have an increased risk of presenting with plasmatic LPS levels [116]. The increase in the number of Gram-negative bacteria in the gut microbiota, the reduction in gut mucosal integrity, and the consumption of high-fat diets increase plasmatic lipopolysaccharide levels [116]. Hence, it is therefore plausible to conclude that the type of diet that patients consume may modulate the composition of the intestinal microbiome that could ultimately improve intestinal barrier mucosal integrity and decrease the occurrence of endotoxemia and its postprandial inflammatory effects. This combined activity leads to adequate insulin signaling [116]. The authors note that there are few studies that have evaluated the influence of nutrients and/or specific food types on metabolic endotoxemia [116].
In a recent study linked to endotoxemia, Cox and colleagues [118] investigated the relationships that can exist between the intestinal microbiome, intestinal permeability (i.e., the intestinal epithelial barrier disruption), and inflammatory responses as multi-factorial risk factors for obesity-associated metabolic diseases. The study aimed to investigate the associations between intestinal epithelial cell permeability and T2DM [118].
The primary outcome data demonstrated that there were differences in the measures of intestinal epithelial cell barrier permeability between individuals with and without T2DM [118]. The authors suggested that using intestinal epithelial cell barrier permeability measures as a tool for predicting T2DM risk has biological plausibility. Consequently, a recent study used plasma zonulin levels (zonulin is a protein modulator of intercellular tight junctions in the intestines) as a non-invasive biomarker of intestinal permeability in women diagnosed with gestational diabetes [119]. Although further studies are warranted, this initial study showed that there was a positive correlation between plasma zonulin levels and body mass index, creatinine, fasting plasma glucose, baseline, first hour, and two-hour glucose levels and the OGTT, haemoglobin A1C (HbA1C), homeostatic model assessment for insulin resistance (HOMA-IR), and alanine aminotransferase (ALT) levels. The authors suggested that the findings could promote zonulin as a non-invasive biomarker involved in the pathogenesis of gestational diabetes [119].
Butyrate, a 4-carbon SCFA produced by intestinal bacteria [120] via glycolysis during the intestinal microbial fermentation of an undigested carbohydrate, has been shown to be a key facilitator in the assembly of the intestinal epithelial barrier [121]. Butyrate exhibits numerous biological activities in different experimental settings, including energy homeostasis, glucose and lipid metabolism, inflammation, redox and neural signaling, and epigenetic modulation [122]. Importantly, butyrate has been reported to down-regulate inflammation by inhibiting the growth of pathobionts, increasing mucosal barrier integrity, encouraging obligate anaerobic bacterial dominance, and decreasing oxygen availability in the gut [122].
Furthermore, butyrate can decrease excessive inflammation through the modulation of immune cells, e.g., by increasing the functionalities of M2 macrophages and regulatory T cells and inhibiting infiltration by neutrophils [122]. Therefore, bacterial species known to be butyrate-producers or dietary components that will enhance the molar ratio of butyrate production are considered as important in maintaining an effective gut barrier and improving glucose metabolism and insulin resistance [123].

5. Discussion

In this narrative review, we have focused on the adjuvant role that functional foods such as prebiotics may provide to the intestinal microbiome to improve metabolic dispositions in T2DM. In addition, while the dietary approach to modulating the intestinal microbiome with the administration of probiotics has a long history, there has always been probiotic survivability and viability issues with the administration of various probiotic formulations that have been difficult to ascertain.
We have previously investigated, through clinical studies, that functional foods with prebiotics and probiotics, co-administered with standard medications, were effective for the management of T2DM. These studies confirmed that the co-administration of an oligofructose prebiotic with sulfonylurea [28] and a multi-strain probiotic plus metformin [27] improved glycemic control in patients diagnosed with T2DM.
The intestinal microbiome has been suggested to have a significant impact on transforming T2DM [124]. The review concluded that there was biologically plausible evidence for the gut microbiome’s ability to influence and improve T2DM symptomatology. Moreover, the authors of the review partially demonstrated the benefit that probiotics, prebiotics, and synbiotics may afford to patients diagnosed with pre-diabetes. The conclusion was that any beneficial effects were centred on modulating the abundance of the intestinal microbiome [124].
Animal and human studies investigating Clostridium species that mostly utilize indigestible polysaccharides by principally fermenting carbohydrates, proteins, and organic acids have shown that these bacteria produce SCFAs of acetic acid, propionic acid, and butyric acid, thereby affording numerous benefits to gut health through intestinal actions which can translate to enhanced management of T2DM [125]. Consequently, research continues to report the importance of SCFAs in the gut, such as that attributed to butyrate, a molecule with ubiquitous epigenetic effects on the gut [120,126]. Among the gram-positive anaerobic bacteria, butyrate-producing bacteria are widely distributed in the intestines. Two of the most important groups of intestinal microbes are Faecalibacterium prausnitzii in the Clostridium leptum cluster (i.e., Clostridial cluster IV) and Eubacterium rectale/Roseburia spp. in the Clostridium coccoides (i.e., Clostridial cluster XIVa) cluster of Firmicutes [127]. These groups of intestinal bacteria typically account for approximately 5–10% of the total bacteria that have been detected in the stool samples of healthy adult humans [127]. These groups of intestinal butyrate-producing bacteria are widely distributed across several groups that include the clusters IX, XV, XVI, and XVII [127].The importance of the presence of Bacteroides resides in their capacity to ferment carbohydrates. Members of the taxa Bacteroides and other intestinal bacteria produce a significant mixture of fatty acids, which can be utilized by the host as an energy source [128], and this may have favourable sequelae in T2DM. An early review reported that the dominant intestinal bacteria consist of anaerobes from the taxa Bacteroides, Bifidobacterium, Eubacterium, Lactobacillus, and Streptococci [129]. However, Enterobacteria were cited as being usually found in a lesser abundance [129]. A recent report investigated the universal healthy intestinal microbiome from numerous geographical areas, establishing that one of the abundant clusters, namely Bacteroides, results in dominant bacterial signatures [130].
Furthermore, a cluster of bacteria consisting of Ruminococcus and Blautia were also reported, and the significance of this is that these clusters have significant positive associations with T2DM [25]. Blautia is widely distributed in mammalian faeces and the intestines. As a dominant genus in the intestinal microbiota, there is a significant correlation between its host physiological and metabolic dysfunctions with obesity, diabetes, and various inflammatory and metabolic diseases due to its antibacterial activity against specific commensal microorganisms [131]. In addition, members of the taxa Ruminococcus also have a relevant importance to T2DM, as the members of this taxon breakdown cellulose that produces methane with the accumulation of a reserve of glucose polymers important in glucose homeostasis [25,130]. Reports that show that galactooligosaccharide reduced the abundance of the Ruminococcus taxa in the intestines may constitute a significant clinical step in prescribing specific prebiotic fibers, beneficially improving the glycemic control of T2DM [132] by reducing the abundance of intestinal microbes that promote diabetes. The associated increase in Bacteroidaceae levels in sulfonylurea monotherapy patients taking adjuvant oligofructose prebiotics provides further support to the posit of a beneficial role of this family of bacteria in T2DM [28].
Patients without metabolic diseases, in contrast to studies that have described the effects that prebiotic fibers may have on the composition of the intestinal microbiota, show consistently reported increases in the abundance of Bifidobacterium [133,134,135] and/or Lactobacillus species [135,136]. Intestinal microbial cross-feeding mechanisms in the colon are thought to form the basis of butyrate production [137], a recognised functional characteristic of several colon bacteria that, in part, explains Bifidobacterium competitiveness and butyrate production [137]. In addition, the specificity of polysaccharide use by the colon microbiota may determine diet-induced alterations in the microbiota and consequent advantageous metabolic effects in patients with T2DM. Certainly, supplementation with nondigestible polysaccharides of plant origin is important for the enrichment of the intestinal microbiota with Lactobacillus and Bifidobacterium species, which ferment these compounds into SCFAs [103,138]. Of further importance are the genera Bifidobacterium, Bacteroides, Faecalibacterium, Akkermansia, and Roseburia that have been reported to be significantly and negatively associated with T2DM [25]. Specifically, the genus Faecalibacterium consists of abundant butyric acid-producing bacteria that colonize the human intestines and display an overall anti-inflammatory effect with the potential of re-regulating adverse inflammatory responses in the gut [139], a prevalent adverse factor that is present in patients diagnosed with T2DM. Similarly, Roseburia also shows stable and relatively abundant levels in the gut that show favourable sequelae in glucose metabolism in T2DM. Members of the Roseburia genus likely play a major role in maintaining gut health and immune defence by maintaining regulatory T-cell homeostasis, predominantly through the production of butyrate [140]. Conversely, a recent study investigating butyrate-producing gut bacteria and insulin homeostasis showed that not all butyrate-producing intestinal bacteria can benefit patients diagnosed with T2DM [141]. The authors reported that although most butyrate producers analysed appear to be metabolically beneficial for T2DM, this was not the case for all such gut bacteria. In contrast, Flavonifractor (i.e., Gram-positive bacteria belonging to the Clostridiales order) was associated with lower insulin sensitivity and disposition index and a higher prevalence of dysglycemia [141]. Hence, a biologically plausible posit ensues that clinical measures to treat or prevent T2DM should be targeted to specific butyrate-producing taxa rather than all gut microbiome butyrate producers [141].

6. Conclusions

The clinical use of supplemented fibers, namely oligofructose, with hypoglycemic medications such as sulfonylureas could improve health outcomes in patients diagnosed with pre-T2DM and early-stage T2DM. The intestinal benefits have been attributed to an increased abundance of butyrate-producing intestinal microbes, including F. prauznitzii, that improve intestinal health maintenance (e.g., reducing intestinal epithelial barrier permeability and pro-inflammatory actions in the gut) and immune defence by maintaining regulatory T-cell homeostasis. Moreover, this bacterium has been promoted as a new generation probiotic [142]; therefore, a prebiotic fiber, such as oligofructose, that can enhance the abundance of such beneficial bacteria in the gut could be posited to improve T2DM metabolic dispositions when subsequently combined with sulfonylurea. Furthermore, it is noteworthy that well-balanced glucose homeostasis may require the presence of several specific bacterial species that work together to provide a number of functions, each of which with distinct properties.

Author Contributions

Conceptualization, L.V.; writing—original draft preparation, L.V.; writing—review and editing, N.N.G., G.V., J.D.H. and L.V. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no internal or external funding, and no fee was provided for drafting and finalising the manuscript.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Figure 1 was adapted from a BioRender template, namely “Gut Epithelial Barrier”, with the author of the manuscript adding content [BioRender.com (2020)].

Conflicts of Interest

The authors L.V., N.N.G. and G.V. declare that they have previously published clinical studies in T2DM with probiotics and prebiotics.

References

  1. Senchukova, M.A. Microbiota of the gastrointestinal tract: Friend or foe? World J. Gastroenterol. 2023, 29, 19–42. [Google Scholar] [CrossRef] [PubMed]
  2. Cunningham, A.L.; Stephens, J.W.; Harris, D.A. Gut microbiota influence in type 2 diabetes mellitus (T2DM). Gut Pathog. 2021, 13, 50. [Google Scholar] [CrossRef] [PubMed]
  3. Thursby, E.; Juge, N. Introduction to the human gut microbiota. Biochem. J. 2017, 474, 1823–1836. [Google Scholar] [CrossRef] [PubMed]
  4. Vivarelli, S.; Salemi, R.; Candido, S.; Falzone, L.; Santagati, M.; Stefani, S.; Torino, F.; Banna, G.L.; Tonini, G.; Libra, M. Gut Microbiota and Cancer: From Pathogenesis to Therapy. Cancers 2019, 11, 38. [Google Scholar] [CrossRef] [PubMed]
  5. Al Bander, Z.; Nitert, M.D.; Mousa, A.; Naderpoor, N. The Gut Microbiota and Inflammation: An Overview. Int. J. Environ. Res. Public Health 2020, 17, 7618. [Google Scholar] [CrossRef]
  6. Maldonado Galdeano, C.; Cazorla, S.I.; Lemme Dumit, J.M.; Vélez, E.; Perdigón, G. Beneficial Effects of Probiotic Consumption on the Immune System. Ann. Nutr. Metab. 2019, 74, 115–124. [Google Scholar] [CrossRef] [PubMed]
  7. Lemme-Dumit, J.M.; Polti, M.A.; Perdigón, G.; Galdeano, C.M. Probiotic bacteria cell walls stimulate the activity of the intestinal epithelial cells and macrophage functionality. Benef. Microbes 2018, 9, 153–164. [Google Scholar] [CrossRef]
  8. Wan, L.Y.; Chen, Z.J.; Shah, N.P.; El-Nezami, H. Modulation of Intestinal Epithelial Defense Responses by Probiotic Bacteria. Crit. Rev. Food Sci. Nutr. 2016, 56, 2628–2641. [Google Scholar] [CrossRef]
  9. Chen, Y.; Zhou, J.; Wang, L. Role and Mechanism of Gut Microbiota in Human Disease. Front. Cell. Infect. Microbiol. 2021, 11, 625913. [Google Scholar] [CrossRef]
  10. Ni, J.; Wu, G.D.; Albenberg, L.; Tomov, V.T. Gut microbiota and IBD: Causation or correlation? Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 573–584. [Google Scholar] [CrossRef]
  11. Kim, D.; Zeng, M.Y.; Núñez, G. The interplay between host immune cells and gut microbiota in chronic inflammatory diseases. Exp. Mol. Med. 2017, 49, e339. [Google Scholar] [CrossRef] [PubMed]
  12. Dembiński, A.; Warzecha, Z.; Ceranowicz, P.; Dembiński, M.; Cieszkowski, J.; Gosiewski, T.; Bulanda, M.; Kuśnierz-Cabala, B.; Gałązka, K.; Konturek, P.C. Synergic Interaction of Rifaximin and Mutaflor (Escherichia coli Nissle 1917) in the Treatment of Acetic Acid-Induced Colitis in Rats. Gastroenterol. Res. Pract. 2016, 2016, 3126280. [Google Scholar] [CrossRef]
  13. Clarke, G.; Stilling, R.M.; Kennedy, P.J.; Stanton, C.; Cryan, J.F.; Dinan, T.G. Minireview: Gut microbiota: The neglected endocrine organ. Mol. Endocrinol. 2014, 28, 1221–1238. [Google Scholar] [CrossRef]
  14. Zhou, L.; Li, X.; Ahmed, A.; Wu, D.; Liu, L.; Qiu, J.; Yan, Y.; Jin, M.; Xin, Y. Gut microbe analysis between hyperthyroid and healthy individuals. Curr. Microbiol. 2014, 69, 675–680. [Google Scholar] [CrossRef]
  15. Zhao, F.; Feng, J.; Li, J.; Zhao, L.; Liu, Y.; Chen, H.; Jin, Y.; Zhu, B.; Wei, Y. Alterations of the Gut Microbiota in Hashimoto’s Thyroiditis Patients. Thyroid 2018, 28, 175–186. [Google Scholar] [CrossRef]
  16. Chakera, A.J.; Pearce, S.H.; Vaidya, B. Treatment for primary hypothyroidism: Current approaches and future possibilities. Drug Des. Dev. Ther. 2012, 6, 1–11. [Google Scholar]
  17. Baker, J.M.; Al-Nakkash, L.; Herbst-Kralovetz, M.M. Estrogen-gut microbiome axis: Physiological and clinical implications. Maturitas 2017, 103, 45–53. [Google Scholar] [CrossRef]
  18. Kwa, M.; Plottel, C.S.; Blaser, M.J.; Adams, S. The Intestinal Microbiome and Estrogen Receptor-Positive Female Breast Cancer. J. Natl. Cancer Inst. 2016, 108, djw029. [Google Scholar]
  19. Yano, J.M.; Yu, K.; Donaldson, G.P.; Shastri, G.G.; Ann, P.; Ma, L.; Nagler, C.R.; Ismagilov, R.F.; Mazmanian, S.K.; Hsiao, E.Y. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell 2015, 161, 264–276. [Google Scholar] [CrossRef]
  20. Moreira, C.G.; Russell, R.; Mishra, A.A.; Narayanan, S.; Ritchie, J.M.; Waldor, M.K.; Curtis, M.M.; Winter, S.E.; Weinshenker, D.; Sperandio, V. Bacterial Adrenergic Sensors Regulate Virulence of Enteric Pathogens in the Gut. mBio 2016, 7, djw029. [Google Scholar] [CrossRef]
  21. Camilleri, M. Leaky gut: Mechanisms, measurement and clinical implications in humans. Gut 2019, 68, 1516–1526. [Google Scholar] [CrossRef]
  22. Usuda, H.; Okamoto, T.; Wada, K. Leaky Gut: Effect of Dietary Fiber and Fats on Microbiome and Intestinal Barrier. Int. J. Mol. Sci. 2021, 22, 7613. [Google Scholar] [CrossRef]
  23. Jaworek, J.; Tudek, B.; Kowalczyk, P.; Kot, M.; Szklarczyk, J.; Leja-Szpak, A.; Pierzchalski, P.; Bonior, J.; Dembiński, A.; Ceranowicz, P.; et al. Effect of Endotoxemia in Suckling Rats on Pancreatic Integrity and Exocrine Function in Adults: A Review Report. Gastroenterol. Res. Pract. 2018, 2018, 6915059. [Google Scholar] [CrossRef]
  24. Sikalidis, A.K.; Maykish, A. The Gut Microbiome and Type 2 Diabetes Mellitus: Discussing a Complex Relationship. Biomedicines 2020, 8, 8. [Google Scholar] [CrossRef]
  25. Gurung, M.; Li, Z.; You, H.; Rodrigues, R.; Jump, D.B.; Morgun, A.; Shulzhenko, N. Role of gut microbiota in type 2 diabetes pathophysiology. EBioMedicine 2020, 51, 102590. [Google Scholar] [CrossRef]
  26. Ojo, O.; Wang, X.; Ojo, O.O.; Brooke, J.; Jiang, Y.; Dong, Q.; Thompson, T. The Effect of Prebiotics and Oral Anti-Diabetic Agents on Gut Microbiome in Patients with Type 2 Diabetes: A Systematic Review and Network Meta-Analysis of Randomised Controlled Trials. Nutrients 2022, 14, 5139. [Google Scholar] [CrossRef]
  27. Palacios, T.; Vitetta, L.; Coulson, S.; Madigan, C.D.; Lam, Y.Y.; Manuel, R.; Briskey, D.; Hendy, C.; Kim, J.N.; Ishoey, T.; et al. Targeting the Intestinal Microbiota to Prevent Type 2 Diabetes and Enhance the Effect of Metformin on Glycaemia: A Randomised Controlled Pilot Study. Nutrients 2020, 12, 2041. [Google Scholar] [CrossRef]
  28. Gorgani, N.N.; Kim, K.H.S.; Free, W.L.; Afkham, M.; Henson, J.D.; Shamanna, P.; Ajdari, S.; Kahn, C.R.; Hirst, T.R.; Barnett, A.H.; et al. OZ101, an oligofructose prebiotic, may prolong sulphonylurea efficacy in patients with type 2 diabetes: A pilot study. J. Curr. Med. Res. Opin. 2022, 5, 1235–1251. [Google Scholar]
  29. Santos-Buelga, C.; González-Paramás, A.M.; Oludemi, T.; Ayuda-Durán, B.; González-Manzano, S. Plant phenolics as functional food ingredients. Adv. Food Nutr. Res. 2019, 90, 183–257. [Google Scholar]
  30. Walker, A.W.; Lawley, T.D. Therapeutic modulation of intestinal dysbiosis. Pharmacol. Res. 2013, 69, 75–86. [Google Scholar] [CrossRef]
  31. Tanase, D.M.; Gosav, E.M.; Costea, C.F.; Ciocoiu, M.; Lacatusu, C.M.; Maranduca, M.A.; Ouatu, A.; Floria, M. The Intricate Relationship between Type 2 Diabetes Mellitus (T2DM), Insulin Resistance (IR), and Nonalcoholic Fatty Liver Disease (NAFLD). J. Diabetes Res. 2020, 2020, 3920196. [Google Scholar] [CrossRef] [PubMed]
  32. Solis-Herrera, C.; Triplitt, C.; Cersosimo, E.; DeFronzo, R.A. Pathogenesis of Type 2 Diabetes Mellitus. In Endotext; Feingold, K.R., Anawalt, B., Blackman, M.R., Boyce, A., Chrousos, G., Corpas, E., Eds.; Copyright © 2000–2023; MDText.com, Inc.: South Dartmouth, MA, USA, 2000. [Google Scholar]
  33. Ağagündüz, D.; Icer, M.A.; Yesildemir, O.; Koçak, T.; Kocyigit, E.; Capasso, R. The roles of dietary lipids and lipidomics in gut-brain axis in type 2 diabetes mellitus. J. Transl. Med. 2023, 21, 240. [Google Scholar] [CrossRef]
  34. McClain, D.A. Glucose Toxicity. In Encyclopedia of Endocrine Diseases; Martini, L., Ed.; Elsevier Inc.: London, UK, 2004. [Google Scholar]
  35. Filippello, A.; Di Mauro, S.; Scamporrino, A.; Malaguarnera, R.; Torrisi, S.A.; Leggio, G.M.; Di Pino, A.; Scicali, R.; Purrello, F.; Piro, S. High Glucose Exposure Impairs L-Cell Differentiation in Intestinal Organoids: Molecular Mechanisms and Clinical Implications. Int. J. Mol. Sci. 2021, 22, 6660. [Google Scholar] [CrossRef] [PubMed]
  36. Zhang, X.; Ha, S.; Lau, H.C.; Yu, J. Excess body weight: Novel insights into its roles in obesity comorbidities. Semin. Cancer Biol. 2023, 92, 16–27. [Google Scholar] [CrossRef] [PubMed]
  37. Ley, R.E.; Turnbaugh, P.J.; Klein, S.; Gordon, J.I. Microbial ecology: Human gut microbes associated with obesity. Nature 2006, 444, 1022–1023. [Google Scholar] [CrossRef]
  38. Li, D.; Li, Y.; Yang, S.; Lu, J.; Jin, X.; Wu, M. Diet-gut microbiota-epigenetics in metabolic diseases: From mechanisms to therapeutics. Biomed. Pharmacother. 2022, 153, 113290. [Google Scholar] [CrossRef]
  39. Zhou, Z.; Sun, B.; Yu, D.; Zhu, C. Gut Microbiota: An Important Player in Type 2 Diabetes Mellitus. Front. Cell. Infect. Microbiol. 2022, 12, 834485. [Google Scholar] [CrossRef]
  40. Stanimirovic, J.; Radovanovic, J.; Banjac, K.; Obradovic, M.; Essack, M.; Zafirovic, S.; Gluvic, Z.; Gojobori, T.; Isenovic, E.R. Role of C-Reactive Protein in Diabetic Inflammation. Mediat. Inflamm. 2022, 2022, 3706508. [Google Scholar] [CrossRef]
  41. Kahn, S.E.; Cooper, M.E.; Del Prato, S. Pathophysiology and treatment of type 2 diabetes: Perspectives on the past, present, and future. Lancet 2014, 383, 1068–1083. [Google Scholar] [CrossRef]
  42. Tilg, H.; Moschen, A.R. Microbiota and diabetes: An evolving relationship. Gut 2014, 63, 1513–1521. [Google Scholar] [CrossRef]
  43. Gordon, S. Elie Metchnikoff, the Man and the Myth. J. Innate Immun. 2016, 8, 223–227. [Google Scholar] [CrossRef] [PubMed]
  44. Jiang, H.; Cai, M.; Shen, B.; Wang, Q.; Zhang, T.; Zhou, X. Synbiotics and Gut Microbiota: New Perspectives in the Treatment of Type 2 Diabetes Mellitus. Foods 2022, 11, 2438. [Google Scholar] [CrossRef]
  45. Zhang, Q.; Hu, N. Effects of Metformin on the Gut Microbiota in Obesity and Type 2 Diabetes Mellitus. Diabetes Metab. Syndr. Obes. 2020, 13, 5003–5014. [Google Scholar] [CrossRef]
  46. Belorkar, S.A.; Gupta, A.K. Oligosaccharides: A boon from nature’s desk. AMB Express 2016, 6, 82. [Google Scholar] [CrossRef] [PubMed]
  47. Dehghan, P.; Pourghassem Gargari, B.; Asghari Jafar-abadi, M. Oligofructose-enriched inulin improves some inflammatory markers and metabolic endotoxemia in women with type 2 diabetes mellitus: A randomized controlled clinical trial. Nutrition 2014, 30, 418–423. [Google Scholar] [CrossRef] [PubMed]
  48. Paul, P.; Kaul, R.; Abdellatif, B.; Arabi, M.; Upadhyay, R.; Saliba, R.; Sebah, M.; Chaari, A. The Promising Role of Microbiome Therapy on Biomarkers of Inflammation and Oxidative Stress in Type 2 Diabetes: A Systematic and Narrative Review. Front. Nutr. 2022, 9, 906243. [Google Scholar] [CrossRef]
  49. An, R.; Zong, A.; Chen, S.; Xu, R.; Zhang, R.; Jiang, W.; Liu, L.; Du, F.; Zhang, H.; Xu, T. Effects of oligosaccharides on the markers of glycemic control: A systematic review and meta-analysis of randomized controlled trials. Food Funct. 2022, 13, 8766–8782. [Google Scholar] [CrossRef]
  50. Slavin, J. Fiber and prebiotics: Mechanisms and health benefits. Nutrients 2013, 5, 1417–1435. [Google Scholar] [CrossRef]
  51. Rezende, E.S.V.; Lima, G.C.; Naves, M.M.V. Dietary fibers as beneficial microbiota modulators: A proposed classification by prebiotic categories. Nutrition 2021, 89, 111217. [Google Scholar] [CrossRef]
  52. Davani-Davari, D.; Negahdaripour, M.; Karimzadeh, I.; Seifan, M.; Mohkam, M.; Masoumi, S.J.; Berenjian, A.; Ghasemi, Y. Prebiotics: Definition, Types, Sources, Mechanisms, and Clinical Applications. Foods 2019, 8, 92. [Google Scholar] [CrossRef]
  53. Chavan, A.R.; Singh, A.K.; Gupta, R.K.; Nakhate, S.P.; Poddar, B.J.; Gujar, V.V.; Purohit, H.J.; Khardenavis, A.A. Recent trends in the biotechnology of functional non-digestible oligosaccharides with prebiotic potential. Biotechnol. Genet. Eng. Rev. 2023, 1–46. [Google Scholar] [CrossRef] [PubMed]
  54. Snelson, M.; de Pasquale, C.; Ekinci, E.I.; Coughlan, M.T. Gut microbiome, prebiotics, intestinal permeability and diabetes complications. Best Pract. Res. Clin. Endocrinol. Metab. 2021, 35, 101507. [Google Scholar] [CrossRef] [PubMed]
  55. Enam, F.; Mansell, T.J. Prebiotics: Tools to manipulate the gut microbiome and metabolome. J. Ind. Microbiol. Biotechnol. 2019, 46, 1445–1459. [Google Scholar] [CrossRef] [PubMed]
  56. Kong, C.; Faas, M.M.; de Vos, P.; Akkerman, R. Impact of dietary fibers in infant formulas on gut microbiota and the intestinal immune barrier. Food Funct. 2020, 11, 9445–9467. [Google Scholar] [CrossRef] [PubMed]
  57. Akkerman, R.; Faas, M.M.; de Vos, P. Non-digestible carbohydrates in infant formula as substitution for human milk oligosaccharide functions: Effects on microbiota and gut maturation. Crit. Rev. Food Sci. Nutr. 2019, 59, 1486–1497. [Google Scholar] [CrossRef]
  58. Paone, P.; Suriano, F.; Jian, C.; Korpela, K.; Delzenne, N.M.; Van Hul, M.; Salonen, A.; Cani, P.D. Prebiotic oligofructose protects against high-fat diet-induced obesity by changing the gut microbiota, intestinal mucus production, glycosylation and secretion. Gut Microbes 2022, 14, 2152307. [Google Scholar] [CrossRef]
  59. Liu, T.; Sun, Z.; Yang, Z.; Qiao, X. Microbiota-derived short-chain fatty acids and modulation of host-derived peptides formation: Focused on host defense peptides. Biomed. Pharmacother. 2023, 162, 114586. [Google Scholar] [CrossRef]
  60. Golpour, F.; Abbasi-Alaei, M.; Babaei, F.; Mirzababaei, M.; Parvardeh, S.; Mohammadi, G.; Nassiri-Asl, M. Short chain fatty acids, a possible treatment option for autoimmune diseases. Biomed. Pharmacother. 2023, 163, 114763. [Google Scholar] [CrossRef]
  61. Broekaert, W.F.; Courtin, C.M.; Verbeke, K.; Van de Wiele, T.; Verstraete, W.; Delcour, J.A. Prebiotic and other health-related effects of cereal-derived arabinoxylans, arabinoxylan-oligosaccharides, and xylooligosaccharides. Crit. Rev. Food Sci. Nutr. 2011, 51, 178–194. [Google Scholar] [CrossRef]
  62. Deleu, S.; Machiels, K.; Raes, J.; Verbeke, K.; Vermeire, S. Short chain fatty acids and its producing organisms: An overlooked therapy for IBD? EBioMedicine 2021, 66, 103293. [Google Scholar] [CrossRef]
  63. Wong, J.M.; de Souza, R.; Kendall, C.W.; Emam, A.; Jenkins, D.J. Colonic health: Fermentation and short chain fatty acids. J. Clin. Gastroenterol. 2006, 40, 235–243. [Google Scholar] [CrossRef]
  64. Bindels, L.B.; Delzenne, N.M.; Cani, P.D.; Walter, J. Towards a more comprehensive concept for prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2015, 12, 303–310. [Google Scholar] [CrossRef] [PubMed]
  65. Rivière, A.; Selak, M.; Lantin, D.; Leroy, F.; De Vuyst, L. Bifidobacteria and Butyrate-Producing Colon Bacteria: Importance and Strategies for Their Stimulation in the Human Gut. Front. Microbiol. 2016, 7, 979. [Google Scholar] [CrossRef] [PubMed]
  66. Peterson, C.T.; Perez Santiago, J.; Iablokov, S.N.; Chopra, D.; Rodionov, D.A.; Peterson, S.N. Short-Chain Fatty Acids Modulate Healthy Gut Microbiota Composition and Functional Potential. Curr. Microbiol. 2022, 79, 128. [Google Scholar] [CrossRef] [PubMed]
  67. Toporovski, M.S.; de Morais, M.B.; Abuhab, A.; Crippa Júnior, M.A. Effect of Polydextrose/Fructooligosaccharide Mixture on Constipation Symptoms in Children Aged 4 to 8 Years. Nutrients 2021, 13, 1634. [Google Scholar] [CrossRef]
  68. Hopkins, M.J.; Englyst, H.N.; Macfarlane, S.; Furrie, E.; Macfarlane, G.T.; McBain, A.J. Degradation of cross-linked and non-cross-linked arabinoxylans by the intestinal microbiota in children. Appl Environ. Microbiol. 2003, 69, 6354–6360. [Google Scholar] [CrossRef]
  69. Kleessen, B.; Sykura, B.; Zunft, H.J.; Blaut, M. Effects of inulin and lactose on fecal microflora, microbial activity, and bowel habit in elderly constipated persons. Am. J. Clin. Nutr. 1997, 65, 1397–1402. [Google Scholar] [CrossRef] [PubMed]
  70. Tuohy, K.M.; Finlay, R.K.; Wynne, A.G.; Gibson, G.R. Human Volunteer Study on the Prebiotic Effects of HP-Inulin—Faecal Bacteria Enumerated Using Fluorescent In Situ Hybridisation (FISH). Anaerobe 2001, 7, 113–118. [Google Scholar] [CrossRef]
  71. Davis, L.M.; Martínez, I.; Walter, J.; Goin, C.; Hutkins, R.W. Barcoded pyrosequencing reveals that consumption of galactooligosaccharides results in a highly specific bifidogenic response in humans. PLoS ONE 2011, 6, e25200. [Google Scholar] [CrossRef]
  72. Hooda, S.; Boler, B.M.; Serao, M.C.; Brulc, J.M.; Staeger, M.A.; Boileau, T.W.; Dowd, S.E.; Fahey, G.C., Jr.; Swanson, K.S. 454 pyrosequencing reveals a shift in fecal microbiota of healthy adult men consuming polydextrose or soluble corn fiber. J. Nutr. 2012, 142, 1259–1265. [Google Scholar] [CrossRef]
  73. Costabile, A.; Fava, F.; Röytiö, H.; Forssten, S.D.; Olli, K.; Klievink, J.; Rowland, I.R.; Ouwehand, A.C.; Rastall, R.A.; Gibson, G.R.; et al. Impact of polydextrose on the faecal microbiota: A double-blind, crossover, placebo-controlled feeding study in healthy human subjects. Br. J. Nutr. 2012, 108, 471–481. [Google Scholar] [CrossRef]
  74. Ballongue, J.; Schumann, C.; Quignon, P. Effects of lactulose and lactitol on colonic microflora and enzymatic activity. Scand. J. Gastroenterol. Suppl. 1997, 222, 41–44. [Google Scholar] [CrossRef]
  75. Elison, E.; Vigsnaes, L.K.; Rindom Krogsgaard, L.; Rasmussen, J.; Sørensen, N.; McConnell, B.; Hennet, T.; Sommer, M.O.; Bytzer, P. Oral supplementation of healthy adults with 2′-O-fucosyllactose and lacto-N-neotetraose is well tolerated and shifts the intestinal microbiota. Br. J. Nutr. 2016, 116, 1356–1368. [Google Scholar] [CrossRef]
  76. Windey, K.; De Preter, V.; Huys, G.; Broekaert, W.F.; Delcour, J.A.; Louat, T.; Herman, J.; Verbeke, K. Wheat bran extract alters colonic fermentation and microbial composition, but does not affect faecal water toxicity: A randomised controlled trial in healthy subjects. Br. J. Nutr. 2015, 113, 225–238. [Google Scholar] [CrossRef]
  77. Holscher, H.D.; Caporaso, J.G.; Hooda, S.; Brulc, J.M.; Fahey, G.C., Jr.; Swanson, K.S. Fiber supplementation influences phylogenetic structure and functional capacity of the human intestinal microbiome: Follow-up of a randomized controlled trial. Am. J. Clin. Nutr. 2015, 101, 55–64. [Google Scholar] [CrossRef]
  78. Whisner, C.M.; Martin, B.R.; Nakatsu, C.H.; Story, J.A.; MacDonald-Clarke, C.J.; McCabe, L.D.; McCabe, G.P.; Weaver, C.M. Soluble Corn Fiber Increases Calcium Absorption Associated with Shifts in the Gut Microbiome: A Randomized Dose-Response Trial in Free-Living Pubertal Females. J. Nutr. 2016, 146, 1298–1306. [Google Scholar] [CrossRef]
  79. Finegold, S.M.; Li, Z.; Summanen, P.H.; Downes, J.; Thames, G.; Corbett, K.; Dowd, S.; Krak, M.; Heber, D. Xylooligosaccharide increases bifidobacteria but not lactobacilli in human gut microbiota. Food Funct. 2014, 5, 436–445. [Google Scholar] [CrossRef]
  80. Chung, Y.C.; Hsu, C.K.; Ko, C.Y.; Chan, Y.C. Dietary intake of xylooligosaccharides improves the intestinal microbiota, fecal moisture, and pH value in the elderly. Nutr Res. 2007, 27, 756–761. [Google Scholar] [CrossRef]
  81. Vandeputte, D.; Falony, G.; Vieira-Silva, S.; Wang, J.; Sailer, M.; Theis, S.; Verbeke, K.; Raes, J. Prebiotic inulin-type fructans induce specific changes in the human gut microbiota. Gut 2017, 66, 1968–1974. [Google Scholar] [CrossRef]
  82. Deehan, E.C.; Yang, C.; Perez-Muñoz, M.E.; Nguyen, N.K.; Cheng, C.C.; Triador, L.; Zhang, Z.; Bakal, J.A.; Walter, J. Precision Microbiome Modulation with Discrete Dietary Fiber Structures Directs Short-Chain Fatty Acid Production. Cell Host Microbe 2020, 27, 389–404.e386. [Google Scholar] [CrossRef]
  83. Havenaar, R. Intestinal health functions of colonic microbial metabolites: A review. Benef. Microbes 2011, 2, 103–114. [Google Scholar] [CrossRef] [PubMed]
  84. Litvak, Y.; Byndloss, M.X.; Bäumler, A.J. Colonocyte metabolism shapes the gut microbiota. Science 2018, 362, eaat9076. [Google Scholar] [CrossRef] [PubMed]
  85. Peng, L.; Li, Z.R.; Green, R.S.; Holzman, I.R.; Lin, J. Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers. J. Nutr. 2009, 139, 1619–1625. [Google Scholar] [CrossRef] [PubMed]
  86. Hardy, H.; Harris, J.; Lyon, E.; Beal, J.; Foey, A.D. Probiotics, prebiotics and immunomodulation of gut mucosal defences: Homeostasis and immunopathology. Nutrients 2013, 5, 1869–1912. [Google Scholar] [CrossRef]
  87. Liu, J.L.; Segovia, I.; Yuan, X.L.; Gao, Z.H. Controversial Roles of Gut Microbiota-Derived Short-Chain Fatty Acids (SCFAs) on Pancreatic β-Cell Growth and Insulin Secretion. Int. J. Mol. Sci. 2020, 21, 910. [Google Scholar] [CrossRef]
  88. Sunkara, L.T.; Jiang, W.; Zhang, G. Modulation of antimicrobial host defense peptide gene expression by free fatty acids. PLoS ONE 2012, 7, e49558. [Google Scholar] [CrossRef]
  89. Kobayashi, M.; Mikami, D.; Kimura, H.; Kamiyama, K.; Morikawa, Y.; Yokoi, S.; Kasuno, K.; Takahashi, N.; Taniguchi, T.; Iwano, M. Short-chain fatty acids, GPR41 and GPR43 ligands, inhibit TNF-α-induced MCP-1 expression by modulating p38 and JNK signaling pathways in human renal cortical epithelial cells. Biochem. Biophys. Res. Commun. 2017, 486, 499–505. [Google Scholar] [CrossRef]
  90. Walker, J.A.; McKenzie, A.N.J. T(H)2 cell development and function. Nat. Rev. Immunol. 2018, 18, 121–133. [Google Scholar] [CrossRef]
  91. Yan, J.; Takakura, A.; Zandi-Nejad, K.; Charles, J.F. Mechanisms of gut microbiota-mediated bone remodeling. Gut Microbes 2018, 9, 84–92. [Google Scholar] [CrossRef]
  92. Garde, S.; Chodisetti, P.K.; Reddy, M. Peptidoglycan: Structure, Synthesis, and Regulation. EcoSal Plus 2021, 9, 2. [Google Scholar] [CrossRef]
  93. Wolf, A.J.; Underhill, D.M. Peptidoglycan recognition by the innate immune system. Nat. Rev. Immunol. 2018, 18, 243–254. [Google Scholar] [CrossRef] [PubMed]
  94. Dzidic, M.; Boix-Amorós, A.; Selma-Royo, M.; Mira, A.; Collado, M.C. Gut Microbiota and Mucosal Immunity in the Neonate. Med. Sci. 2018, 6, 56. [Google Scholar] [CrossRef] [PubMed]
  95. Rawi, M.H.; Zaman, S.A.; Pa’ee, K.F.; Leong, S.S.; Sarbini, S.R. Prebiotics metabolism by gut-isolated probiotics. J. Food Sci. Technol. 2020, 57, 2786–2799. [Google Scholar] [CrossRef] [PubMed]
  96. Roy, S.; Dhaneshwar, S. Role of prebiotics, probiotics, and synbiotics in management of inflammatory bowel disease: Current perspectives. World J. Gastroenterol. 2023, 29, 2078–2100. [Google Scholar] [CrossRef]
  97. Zhang, X.F.; Guan, X.X.; Tang, Y.J.; Sun, J.F.; Wang, X.K.; Wang, W.D.; Fan, J.M. Clinical effects and gut microbiota changes of using probiotics, prebiotics or synbiotics in inflammatory bowel disease: A systematic review and meta-analysis. Eur. J. Nutr. 2021, 60, 2855–2875. [Google Scholar] [CrossRef]
  98. Liu, N.; Wang, H.; Yang, Z.; Zhao, K.; Li, S.; He, N. The role of functional oligosaccharides as prebiotics in ulcerative colitis. Food Funct. 2022, 13, 6875–6893. [Google Scholar] [CrossRef]
  99. Hartemink, R.; Quataert, M.C.; van Laere, K.M.; Nout, M.J.; Rombouts, F.M. Degradation and fermentation of fructo-oligosaccharides by oral streptococci. J. Appl. Bacteriol. 1995, 79, 551–557. [Google Scholar] [CrossRef]
  100. Hartemink, R.; Van Laere, K.M.; Rombouts, F.M. Growth of enterobacteria on fructo-oligosaccharides. J. Appl. Microbiol. 1997, 83, 367–374. [Google Scholar] [CrossRef]
  101. Van Laere, K.M.; Hartemink, R.; Bosveld, M.; Schols, H.A.; Voragen, A.G. Fermentation of plant cell wall derived polysaccharides and their corresponding oligosaccharides by intestinal bacteria. J. Agric. Food Chem. 2000, 48, 1644–1652. [Google Scholar] [CrossRef]
  102. Li, K.-K.; Tian, P.-J.; Wang, S.-D.; Lei, P.; Qu, L.; Huang, J.-P.; Shan, Y.-J.; Li, B.-L. Targeting gut microbiota: Lactobacillus alleviated type 2 diabetes via inhibiting LPS secretion and activating GPR43 pathway. J. Funct. Foods 2017, 561–570. [Google Scholar] [CrossRef]
  103. Markowiak-Kopeć, P.; Śliżewska, K. The Effect of Probiotics on the Production of Short-Chain Fatty Acids by Human Intestinal Microbiome. Nutrients 2020, 12, 1107. [Google Scholar] [CrossRef] [PubMed]
  104. Bodinham, C.L.; Smith, L.; Thomas, E.L.; Bell, J.D.; Swann, J.R.; Costabile, A.; Russell-Jones, D.; Umpleby, A.M.; Robertson, M.D. Efficacy of increased resistant starch consumption in human type 2 diabetes. Endocr. Connect. 2014, 3, 75–84. [Google Scholar] [CrossRef] [PubMed]
  105. Lu, Z.X.; Walker, K.Z.; Muir, J.G.; O’Dea, K. Arabinoxylan fibre improves metabolic control in people with Type II diabetes. Eur. J. Clin. Nutr. 2004, 58, 621–628. [Google Scholar] [CrossRef]
  106. Dixon, S.A.; Mishra, S.; Dietsche, K.B.; Jain, S.; Mabundo, L.; Stagliano, M.; Krenek, A.; Courville, A.; Yang, S.; Turner, S.A.; et al. The effects of prebiotics on gastrointestinal side effects of metformin in youth: A pilot randomized control trial in youth-onset type 2 diabetes. Front. Endocrinol. 2023, 14, 1125187. [Google Scholar] [CrossRef] [PubMed]
  107. Saul, D.; Kosinsky, R.L. Epigenetics of Aging and Aging-Associated Diseases. Int. J. Mol. Sci. 2021, 22, 401. [Google Scholar] [CrossRef]
  108. Ling, C.; Rönn, T. Epigenetics in Human Obesity and Type 2 Diabetes. Cell Metab. 2019, 29, 1028–1044. [Google Scholar] [CrossRef]
  109. Ling, C. Epigenetic regulation of insulin action and secretion–role in the pathogenesis of type 2 diabetes. J. Intern. Med. 2020, 288, 158–167. [Google Scholar] [CrossRef]
  110. Larsen, N.; Vogensen, F.K.; van den Berg, F.W.; Nielsen, D.S.; Andreasen, A.S.; Pedersen, B.K.; Al-Soud, W.A.; Sørensen, S.J.; Hansen, L.H.; Jakobsen, M. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS ONE 2010, 5, e9085. [Google Scholar] [CrossRef]
  111. Qin, J.; Li, Y.; Cai, Z.; Li, S.; Zhu, J.; Zhang, F.; Liang, S.; Zhang, W.; Guan, Y.; Shen, D.; et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012, 490, 55–60. [Google Scholar] [CrossRef]
  112. Karlsson, F.H.; Tremaroli, V.; Nookaew, I.; Bergström, G.; Behre, C.J.; Fagerberg, B.; Nielsen, J.; Bäckhed, F. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature 2013, 498, 99–103. [Google Scholar] [CrossRef]
  113. Artasensi, A.; Mazzolari, A.; Pedretti, A.; Vistoli, G.; Fumagalli, L. Obesity and Type 2 Diabetes: Adiposopathy as a Triggering Factor and Therapeutic Options. Molecules 2023, 28, 3094. [Google Scholar] [CrossRef] [PubMed]
  114. Wu, J.; Yang, K.; Fan, H.; Wei, M.; Xiong, Q. Targeting the gut microbiota and its metabolites for type 2 diabetes mellitus. Front. Endocrinol. 2023, 14, 1114424. [Google Scholar] [CrossRef] [PubMed]
  115. Gomes, J.M.G.; Costa, J.A.; Alfenas, R.C.G. Metabolic endotoxemia and diabetes mellitus: A systematic review. Metabolism 2017, 68, 133–144. [Google Scholar] [CrossRef] [PubMed]
  116. Boroni Moreira, A.P.; de Cássia Gonçalves Alfenas, R. The influence of endotoxemia on the molecular mechanisms of insulin resistance. Nutr. Hosp. 2012, 27, 382–390. [Google Scholar]
  117. Opal, S.M.; Scannon, P.J.; Vincent, J.L.; White, M.; Carroll, S.F.; Palardy, J.E.; Parejo, N.A.; Pribble, J.P.; Lemke, J.H. Relationship between plasma levels of lipopolysaccharide (LPS) and LPS-binding protein in patients with severe sepsis and septic shock. J. Infect. Dis. 1999, 180, 1584–1589. [Google Scholar] [CrossRef]
  118. Cox, A.J.; Zhang, P.; Bowden, D.W.; Devereaux, B.; Davoren, P.M.; Cripps, A.W.; West, N.P. Increased intestinal permeability as a risk factor for type 2 diabetes. Diabetes Metab. 2017, 43, 163–166. [Google Scholar] [CrossRef]
  119. Demir, E.; Ozkan, H.; Seckin, K.D.; Sahtiyancı, B.; Demir, B.; Tabak, O.; Kumbasar, A.; Uzun, H. Plasma Zonulin Levels as a Non-Invasive Biomarker of Intestinal Permeability in Women with Gestational Diabetes Mellitus. Biomolecules 2019, 9, 24. [Google Scholar] [CrossRef]
  120. Vitetta, L.; Bambling, M.; Strodl, E. Probiotics and Commensal Bacteria Metabolites Trigger Epigenetic Changes in the Gut and Influence Beneficial Mood Dispositions. Microorganisms 2023, 11, 1334. [Google Scholar] [CrossRef]
  121. Salvi, P.S.; Cowles, R.A. Butyrate and the Intestinal Epithelium: Modulation of Proliferation and Inflammation in Homeostasis and Disease. Cells 2021, 10, 1775. [Google Scholar] [CrossRef]
  122. Chen, J.; Vitetta, L. The Role of Butyrate in Attenuating Pathobiont-Induced Hyperinflammation. Immune Netw. 2020, 20, e15. [Google Scholar] [CrossRef]
  123. Gueddouri, D.; Caüzac, M.; Fauveau, V.; Benhamed, F.; Charifi, W.; Beaudoin, L.; Rouland, M.; Sicherre, F.; Lehuen, A.; Postic, C.; et al. Insulin resistance per se drives early and reversible dysbiosis-mediated gut barrier impairment and bactericidal dysfunction. Mol. Metab. 2022, 57, 101438. [Google Scholar] [CrossRef] [PubMed]
  124. Li, W.Z.; Stirling, K.; Yang, J.J.; Zhang, L. Gut microbiota and diabetes: From correlation to causality and mechanism. World J. Diabetes. 2020, 11, 293–308. [Google Scholar] [CrossRef]
  125. Guo, P.; Zhang, K.; Ma, X.; He, P. Clostridium species as probiotics: Potentials and challenges. J. Anim. Sci. Biotechnol. 2020, 11, 24. [Google Scholar] [CrossRef] [PubMed]
  126. Berni Canani, R.; Di Costanzo, M.; Leone, L. The epigenetic effects of butyrate: Potential therapeutic implications for clinical practice. Clin. Epigenet. 2012, 4, 4. [Google Scholar] [CrossRef] [PubMed]
  127. Singh, V.; Lee, G.; Son, H.; Koh, H.; Kim, E.S.; Unno, T.; Shin, J.H. Butyrate producers, “The Sentinel of Gut”: Their intestinal significance with and beyond butyrate, and prospective use as microbial therapeutics. Front. Microbiol. 2022, 13, 1103836. [Google Scholar] [CrossRef]
  128. Hooper, L.V.; Midtvedt, T.; Gordon, J.I. How host-microbial interactions shape the nutrient environment of the mammalian intestine. Annu. Rev. Nutr. 2002, 22, 283–307. [Google Scholar] [CrossRef]
  129. Liu, H.; Wang, J.; He, T.; Becker, S.; Zhang, G.; Li, D.; Ma, X. Butyrate: A Double-Edged Sword for Health? Adv. Nutr. 2018, 9, 21–29. [Google Scholar] [CrossRef]
  130. Piquer-Esteban, S.; Ruiz-Ruiz, S.; Arnau, V.; Diaz, W.; Moya, A. Exploring the universal healthy human gut microbiota around the World. Comput. Struct. Biotechnol. J. 2022, 20, 421–433. [Google Scholar] [CrossRef]
  131. Kalyana Chakravarthy, S.; Jayasudha, R.; Sai Prashanthi, G.; Ali, M.H.; Sharma, S.; Tyagi, M.; Shivaji, S. Dysbiosis in the Gut Bacterial Microbiome of Patients with Uveitis, an Inflammatory Disease of the Eye. Indian J. Microbiol. 2018, 58, 457–469. [Google Scholar] [CrossRef]
  132. Liu, F.; Li, P.; Chen, M.; Luo, Y.; Prabhakar, M.; Zheng, H.; He, Y.; Qi, Q.; Long, H.; Zhang, Y.; et al. Fructooligosaccharide (FOS) and Galactooligosaccharide (GOS) Increase Bifidobacterium but Reduce Butyrate Producing Bacteria with Adverse Glycemic Metabolism in healthy young population. Sci. Rep. 2017, 7, 11789. [Google Scholar] [CrossRef]
  133. Azpiroz, F.; Molne, L.; Mendez, S.; Nieto, A.; Manichanh, C.; Mego, M.; Accarino, A.; Santos, J.; Sailer, M.; Theis, S.; et al. Effect of Chicory-derived Inulin on Abdominal Sensations and Bowel Motor Function. J. Clin. Gastroenterol. 2017, 51, 619–625. [Google Scholar] [CrossRef]
  134. Staudacher, H.M.; Whelan, K. Altered gastrointestinal microbiota in irritable bowel syndrome and its modification by diet: Probiotics, prebiotics and the low FODMAP diet. Proc. Nutr. Soc. 2016, 75, 306–318. [Google Scholar] [CrossRef] [PubMed]
  135. Sakai, Y.; Seki, N.; Hamano, K.; Ochi, H.; Abe, F.; Masuda, K.; Iino, H. Prebiotic effect of two grams of lactulose in healthy Japanese women: A randomised, double-blind, placebo-controlled crossover trial. Benef. Microbes 2019, 10, 629–639. [Google Scholar] [CrossRef] [PubMed]
  136. You, S.; Ma, Y.; Yan, B.; Pei, W.; Wu, Q.; Ding, C.; Huang, C. The promotion mechanism of prebiotics for probiotics: A review. Front. Nutr. 2022, 9, 1000517. [Google Scholar] [CrossRef]
  137. De Vuyst, L.; Leroy, F. Cross-feeding between bifidobacteria and butyrate-producing colon bacteria explains bifdobacterial competitiveness, butyrate production, and gas production. Int. J. Food Microbiol. 2011, 149, 73–80. [Google Scholar] [CrossRef] [PubMed]
  138. Horiuchi, H.; Kamikado, K.; Aoki, R.; Suganuma, N.; Nishijima, T.; Nakatani, A.; Kimura, I. Bifidobacterium animalis subsp. lactis GCL2505 modulates host energy metabolism via the short-chain fatty acid receptor GPR43. Sci. Rep. 2020, 10, 4158. [Google Scholar] [CrossRef]
  139. Boyanova, L.; Markovska, R.; Yordanov, D.; Gergova, R.; Hadzhiyski, P. Anaerobes in specific infectious and noninfectious diseases: New developments. Anaerobe 2023, 81, 102714. [Google Scholar] [CrossRef]
  140. Tamanai-Shacoori, Z.; Smida, I.; Bousarghin, L.; Loreal, O.; Meuric, V.; Fong, S.B.; Bonnaure-Mallet, M.; Jolivet-Gougeon, A. Roseburia spp.: A marker of health? Future Microbiol. 2017, 12, 157–170. [Google Scholar] [CrossRef]
  141. Cui, J.; Ramesh, G.; Wu, M.; Jensen, E.T.; Crago, O.; Bertoni, A.G.; Gao, C.; Hoffman, K.L.; Sheridan, P.A.; Wong, K.E.; et al. Butyrate-Producing Bacteria and Insulin Homeostasis: The Microbiome and Insulin Longitudinal Evaluation Study (MILES). Diabetes 2022, 71, 2438–2446. [Google Scholar] [CrossRef]
  142. Cheng, H.L.; Yen, G.C.; Huang, S.C.; Chen, S.C.; Hsu, C.L. The next generation beneficial actions of novel probiotics as potential therapeutic targets and prediction tool for metabolic diseases. J. Food Drug Anal. 2022, 30, 1–10. [Google Scholar] [CrossRef]
Figure 1. Prebiotics increase the concentration of luminal butyrate [85], facilitate tight junction barrier assembly, improve the integrity of the intestinal epithelium that inhibits the translocation of endotoxins (e.g., lipopolysaccharide) and decrease the abundance of Gram-negative bacteria [84]; a large proportion of the of the indigenous bacteria are Gram-negative, which accounts for the high LPS load that the colonic gut can experience [86].
Figure 1. Prebiotics increase the concentration of luminal butyrate [85], facilitate tight junction barrier assembly, improve the integrity of the intestinal epithelium that inhibits the translocation of endotoxins (e.g., lipopolysaccharide) and decrease the abundance of Gram-negative bacteria [84]; a large proportion of the of the indigenous bacteria are Gram-negative, which accounts for the high LPS load that the colonic gut can experience [86].
Biomolecules 13 01307 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Vitetta, L.; Gorgani, N.N.; Vitetta, G.; Henson, J.D. Prebiotics Progress Shifts in the Intestinal Microbiome That Benefits Patients with Type 2 Diabetes Mellitus. Biomolecules 2023, 13, 1307. https://doi.org/10.3390/biom13091307

AMA Style

Vitetta L, Gorgani NN, Vitetta G, Henson JD. Prebiotics Progress Shifts in the Intestinal Microbiome That Benefits Patients with Type 2 Diabetes Mellitus. Biomolecules. 2023; 13(9):1307. https://doi.org/10.3390/biom13091307

Chicago/Turabian Style

Vitetta, Luis, Nick N. Gorgani, Gemma Vitetta, and Jeremy D. Henson. 2023. "Prebiotics Progress Shifts in the Intestinal Microbiome That Benefits Patients with Type 2 Diabetes Mellitus" Biomolecules 13, no. 9: 1307. https://doi.org/10.3390/biom13091307

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop