Next Article in Journal
Diversity of Bacterioplankton and Bacteriobenthos from the Veracruz Reef System, Southwestern Gulf of Mexico
Previous Article in Journal
Taxonomic and Functional Characteristics of the Gill and Gastrointestinal Microbiota and Its Correlation with Intestinal Metabolites in NEW GIFT Strain of Farmed Adult Nile Tilapia (Oreochromis niloticus)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recognizing the Benefits of Pre-/Probiotics in Metabolic Syndrome and Type 2 Diabetes Mellitus Considering the Influence of Akkermansia muciniphila as a Key Gut Bacterium

by
Raluca Anca Corb Aron
1,
Areha Abid
2,
Cosmin Mihai Vesa
1,
Aurelia Cristina Nechifor
3,
Tapan Behl
4,
Timea Claudia Ghitea
5,
Mihai Alexandru Munteanu
6,
Ovidiu Fratila
6,
Felicia Liana Andronie-Cioara
7,
Mirela Marioara Toma
5 and
Simona Bungau
5,*
1
Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
2
Department of Food Science, Faculty of Agricultural and Food Sciences, University of Debrecen, 4032 Debrecen, Hungary
3
Department of Analytical Chemistry, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 011061 Bucharest, Romania
4
Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
5
Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
6
Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
7
Department of Psycho-Neuroscience and Recovery, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
*
Author to whom correspondence should be addressed.
Submission received: 5 March 2021 / Revised: 9 March 2021 / Accepted: 15 March 2021 / Published: 17 March 2021
(This article belongs to the Section Gut Microbiota)

Abstract

:
Metabolic syndrome (MetS) and type 2 diabetes mellitus (T2DM) are diseases that can be influenced by the structure of gut microbiota, whose improvement is often neglected in metabolic pathology. This review highlights the following main aspects: the relationship between probiotics/gut microbes with the pathogenesis of MetS, the particular positive roles of Akkermansia muciniphila supplementation in the onset of MetS, and the interaction between dietary polyphenols (prebiotics) with gut microbiota. Therefore, an extensive and in-depth analysis of the often-neglected correlation between gut microbiota and chronic metabolic diseases was conducted, considering that this topic continues to fascinate and stimulate researchers through the discovery of novel strains and their beneficial properties.

1. Introduction

Metabolic syndrome (MetS) is a combination of risk factors, such as overweight/obesity, hypertension, and imbalances in the metabolism of lipids and carbohydrates. All components of MetS are well-known risk factors when it comes to developing type 2 diabetes mellitus (T2DM), heart diseases, and atherosclerosis. As a consequence of its pathophysiological association with other cardiovascular risks, this chronic condition is one of the main contributors to the prevalence of the disease. As obesity is a precursor of MetS, the maintenance and regulation of MetS will be important in the treatment of obesity by physical activity (e.g., exercises), lifestyle improvements (counseling), caloric restriction menu, reduction of weight by medication, or through weight loss surgery. Strategies such as exercise and behavioral improvement involve firm discipline of the mind and are difficult to follow. In comparison, calorie-limited diets in obese children have shown to be less efficient. The latest pharmacological drugs often determine adverse side-effects and high treatment costs [1].
Several researchers have suggested various nutritional solutions based on probiotic or prebiotic approaches, following a close association between food, gut microbiota, and MetS pathophysiology.
Prebiotics are a peculiar type of dietary fiber with health benefits that not only selectively administer live microbes but are also invoking alterations in the host microbial environment [2]. They can be found in a variety of sources, such are non-digestible oligosaccharides, non-digestible sugars, non-refined wheat, non-refined barley, soy, crude oats, breast milk, and inulin sources (e.g., chicory roots and Jerusalem artichoke) [3]. Prebiotics used as non-digestible food additives have a favorable effect on the host by selectively inducing a small number of colon microorganisms to develop and/or to be enabled [4]. Inulin-based fructose oligomers or galactic oligosaccharides are the majority of prebiotics [5]. Pullulan has long been used for food additives as a possible prebiotic compound [6]. Pullulan is a maltotriosis α-1.6-related polymer and is separated from the Aureobasidium pullulan fungus [7]. It is also considered a non-digestible carbohydrate since it has a high molecular weight and has sluggish hydrolysis of α-amylase and glucoamylase [8]. Pullulan fermented via microbiota has previously been documented to modify the composition of the intestinal microbiota [9]. The FAO and WHO have described probiotics as non-pathogenic living microorganisms, which guarantee the proper health of the host when properly used in foods or food supplements [10].
Probiotics are derived from multiple ways, such as diverse natural habitats, human intestinal microbiota, and food [3,11]. The prophylactic and therapeutic actions of some pre-/probiotics in numerous gut-related disorders are partially mediated via modification or the function of the microbiota. Considering previous definitions, probiotics acts “by contributing to [the host’s] intestinal microbial balance” [12] or “by improving the properties of the indigenous microflora” [13]. Taking into account those mentioned so far, in the current consensual form of the definition of probiotics, their effects are considered to be mediated both by microbiota and by other mechanisms. Thus, probiotic microorganisms can function via numerous pathways, including modulation of immune function; interaction with the resident microbiota; host interface; formation of organic acids, enzymes, or other antimicrobial compounds; and improving the quality of the intestinal barrier, etc.
On the other hand, prebiotics are considered to be substrates through which host microorganisms selectively confer a health benefit, their actions including mineral absorption, defense against pathogens, metabolic effects and regulation of satiety, bowel function, immune modulation, etc. The use of pre-/probiotics has been demonstrated by credible and pertinent evaluations of their effectiveness, but it should be mentioned that not all tested products have been validated. The most important target here can be considered the recommendation of these supplements (clearly based on scientific evidence) only by health professionals [14].
Research has shown that the gut microbiota is one of the host’s most significant environmental influences and establishes dynamic interactions with the hosts over their entire existence. The metabolic role of intestinal microbiota is also crucial to biochemicals in the body that save energy, produce energy-efficient compounds, and produce vitamins and other essential nutrients [15]. In addition, the intestinal microbiology protects its host from pathogenic inflammation and regulates the innate and acquired immunity [16,17]. Researchers have recently reported imbalances in intestinal microbiota, disease sensitivity, immune deficiencies, and, most specifically, obesity and resistance to insulin [18].
Other studies have shown that the prevalence and treatment of diabetes mellitus (DM), in particular T2DM, requires the use of the host microbiome. Dysbiosis has a clear association between T2D and the gut microbiota (GM) of T2D patients with opportunistic pathogens, particularly the butyrate that generates the bacteria, decreasing the number of beneficial microbes. The incoherence of gut microbiota (GM) is considerable. Improving intestinal health can help control diabetic problems and can determine more DM-related health issues from arising [19,20].
Akkermansia muciniphila (A. muciniphila) is a new highly promising probiotic. It colonizes the mucosal layer in the intestines and modulates the basal metabolism. The association between A. muciniphila and obesity is consistent. A number of animal models and human trials have shown the causal positive effect of A. muciniphila therapy on obesity, this probiotic being an enhancer actor in the metabolism of organism and having considerable potential for treating obesity-related metabolism, as well as related to the treatment including newly medicinal agents or compounds [21,22]. In this regard, a good example are the results of Shang et al., illustrating the beneficial effects of Akkermansia on MetS during the treatment of the disease by fucoidan [21].
The purpose of this in-depth analysis, of a topic that may seem exhausted at first glance, is to provide a very complex and complete overview regarding the relationship between probiotics/gut microbes with the pathogenesis of MetS, the roles of A. muciniphila in the onset of MetS, and the interaction between dietary polyphenols (prebiotics) with gut microbiota. The present paper thus consists of extremely concentrated and useful referenced information for any future works in the field. The reason we selected the current subject was the increasing prevalence of MS and its associated diseases, considering also the fact that numerous therapies are not controlled from the point of view of objectives attainment; therefore, additional measures are needed. Furthermore, supplementation with pre-/probiotics is an easy way to give to the patient another modality to control or at least ameliorate his condition and improve his quality of life.

2. Methodology

This review involved an exhaustive research of the literature and included the selection of scientific published data addressing the topic of supplementation with pre-/probiotics in MetS and T2DM. To capture all the details relevant to the research, there was no restriction on the publication date of the selected articles, being included both recent and older works. Moreover, for the purpose mentioned above, the most relevant medical and biological data bases (PubMed/MEDLINE, EMBASE, Science Direct, etc.) for articles published until February 2021 were searched, using the representative keywords (mentioned at the beginning of this paper) in the topic or their combinations, in order to find the main data/results/papers in the field. The flow chart describing the selection process of the articles included in this research is presented in Figure 1. Were excluded the publications that were not on the topic approached in this paper and those that were not in English.

3. The Relationship between Probiotics/Gut Microbes with the Pathogenesis of Metabolic Syndrome

3.1. Intestinal Microbiota and the Link to MetS

The connection between the composition of intestinal microbiome and metabolic diseases, including obesity and diabetes, was seen in recent studies focused on large-scale sequencing of the 16S rRNA, quantitative real-time PCR (qRT-PCR), in situ fluorescing hybridization (FISH), high-throughput technology pyrosequencing, and deoxy-ribonucleic acid (DNA) bar codes. Researchers demonstrated that obesity may cause the intestinal microbiota composition to change both in mice and people. In contrast with their fat counterparts, lean laboratory animals (i.e., rats, mice and pigs) have a larger amount of Bacteroidetes spp., where Firmicutes are prevalent [23,24,25,26,27]. Therefore, in comparison to lean subjects, a decreased ratio of Bacteroidetes/Firmicutes was identified. There is, however, a lack of coherence between studies. Low microbial gene richness has recently been hypothesized as a good marker for MetS [28,29]. In addition, an abnormal gut microbiota is able to induce sub-acute systemic inflammation, insulin tolerance, and increased risk of CVD through bacterial exposure pathways, namely, bacterial lipopolysaccharides (LPS) [30].
Owing to the mixture of genetic and environmental causes, the prevalence of obesity, diabetes, and MetS is attributed to gastrointestinal microbiota. There are several indications that obesity and related intestinal microbiota metabolic disorders are associated with low-grade systemic inflammation [31,32]. The treatment of metabolic disorders and of obesity-related endotoxemia is important in preventing intestinal microbiota dysbiosis and maintaining the function of the epithelial barrier of the intestine [33]. Metabolic endotoxemia is the pathological disorder attributed to low-grade LPS (endotoxin) plasma elevation from the intestines to the bloodstream [34]. Studies have shown that this endotoxin exacerbates chronic metabolic disease pathogenesis and is common in people with DM, dyslipidemia, insulin tolerance, and obesity in subclinical inflammatory processes [35].

3.1.1. Influence of Intestinal Microbiota Dysbiosis

The cause of dysbiosis is related to the loss of usual functions of the microbiota generating many metabolism disorders [36]. A fat-rich diet can also affect the dysbiosis process, resulting in increased serum lipopolysaccharide (LPS) and dysfunctions of the intestinal barrier [37]. In comparison, dysbiosis can aggravate the still unknown pathogenesis of chronic inflammatory disease [38].
Some bacterial cell components have an impact on the immunomodulatory function of the lymphoid tissue in the increased inflammatory phase. Researchers suggest that the translocation and signaling of antigens via the intestinal mucosal barrier are activated by cell membrane components, including peptidoglycans and LPS. A pro-inflammatory function is accomplished by binding LPS and peptidoglycan to toll-like receptor 2 (TLR4) and by activating the immune system in a cascade reaction with the oligomerization domains of nuclear oligotides (NOD) [39,40].
The LPS serum levels are double in obese, diabetic, or extremely fat people, as a result of reduced intestinal barrier resilience, increased development of chylomicron during the digestive phase, and a reduction in the function of the alkaline phosphatasis, which causes the cleavage of this endotoxin. This repeated sensitivity of MetS to serum LPS has also caused MetS association with the innate immune system, assisted by the use of the protein LPS (LBP) and CD14 co-receptor [41,42].
In the intestinal microbiota and the innate immune system, MetS production is interactive. The latest actions are aimed at creating a specific diet to reconstruct and/or strengthen the immune system underlying the intestinal homeostasis of microbiota [43]. In addition, the unique strain properties of gut microbiota have been shown by laboratory models for the attenuation of some chronic infectious immune responses [43].
While the bowel microbiota of adults is constant, food, genotypical/epigenetic combination and immune-metabolic function may determine adjustments. The signaling pathways between the host and microbiome, including different groups of ligands, are used as essential effectors for modification, as stated by Moran and Shanahan in 2014 [44,45]. β-cell syntheses, which control insulin and glucose homeostasis, are enhanced by the involvement of inflammatory biomarkers in diabetes-induced oxidative and terminal stresses [46].
If the structure of the intestinal microbiota becomes more intricate and contagious, more modifications are seen in the macrophage M2 to M1 adiposis environment, which can contribute to the development of MetS [47].

3.1.2. Influence of Dietary Composition

Although the intestinal microbiome responds to major caloric intake changes, numerous experiments suggest that it is most vulnerable to dietary composition [48,49]. In a human trial, volunteers had either a plant-based (cereal, fruit, legumes) or animal-based (meats, eggs, cheeses) diet for five consecutive days. After these five days, their microbial populations underwent major behavioral shifts. Participants whose diet focused on animal products witnessed a bloom of bile-resistant microbes (Alistipes spp., Bilophila spp., and Bacteroides spp.) and a reduction of fiber-fermented bacteria [48]. Additional experiments have shown microbial exposure to seeds, fiber forms, and food contaminants in dietary fats [50,51,52,53].
Particularly interesting are dietary fats, since certain acids have antimicrobial activity, but this feature is calculated by the number of carbons and double C=C-bonds, their position, and orientation [54]. Caesar also found that the proinflammatory Bilophila wadsworthia has bloomed in rats who have been fed a lard diet, while Lactobacillus and A. muciniphila have bloomed in the diet based on fish oil [55]. An increase in the number of A. muciniphila was adversely connected to obesity, T2DM with medication, and elevated BP [56,57,58].
Recently, in overweight/obese insulin-resistant people, the first clinical trial involving oral A. muciniphila supplementation was carried out. The randomized, double-blind combination study was conducted for 3 months and revealed an increase in insulin sensibility and reduction in insulinemia and overall plasma cholesterol. Therefore, it may also be part of the solution, even though the interaction between the diet and the microbiome implies the progression of obesity [59].
An impaired microbiota is further involved in promoting the risk of cardiovascular disease. Studies performed on antibiotic and germ-free mice indicate an important role of gut microbiota in transforming nutrient phosphatidylcholine in trimethylamine N-oxide molecule (TMAO) [60,61]. In this direction, microbes cleave dietary choline through the hepatic flavin mono oxygenize 3 (FMO3) to form TMAO from animal products, which is oxidized, in return, by the liver. The levels of circulating TMAO in patients with atherosclerosis were substantially higher relative to the stable controls [61]. In recent human research, vegan and omnivores who have been confronted with a supplementing choline diet have demonstrated a dose-dependent rise in TMAO in the circulation. After treatment with large spectral antibiotics, the prothrombotic phenotype is absolutely removed, which clearly indicates that the microbiome has an important role in this phenomenon [62].

3.1.3. Influence of Inflammation

Although a consistently low inflammation is not a distinctive cause for MetS, it is a recognized element in the etiopathology of obesity/insulin resistance and is thus closely related to MetS metabolism. The important function of intestinal permeability in persistent low-grade inflammation renders the microbiota as a crucial part of the inflammation in metabolic deficiencies.
Inflammation represents a series of responses of the vascularized tissue to damage or infection. This is a protective survival function, but prolonged sensitivity to stimuli and immune movement in cells can be harmful. The first study of chronic low-degree inflammation with obesity and insulin resistance was documented by Hotamisligl and others in 1993. Rotary adipose tissue tolerance to insulin was achieved by TNF-α overexpression [63,64]. This survey was eventually extended to include obese people, who had elevated TNF-α mRNA as opposed to the controls in obese adipose tissue. Scientists found that metabolic cells, such as adipocytes and hepatocytes, had closely related immune systems and vessels. This proximity provides a continuous communication between the immune response and metabolism [65,66].
In fact, multiple studies found that adipose tissue in obese individuals has increased proinflammatory markers (with expanded adipocytes in mice and humans with macrophage and dead crown like structures of phagocytic macrophages) [67,68].
Inflammatory incidents contribute to an altered metabolism, frequently determined by the experimental usage of high-fat diets; nevertheless, considerable attempts are being made to clarify which causes, aside from a high-fat diet (HFD), are triggers of inflammation [69].
The function of the intracellular endocannabinoid system (ECB) is another interesting gut-centered process that can contribute to low-grade inflammatory dysmetabolism. Obesity increased the ECB levels in adipose tissue and plasma and a strong ECB synthesis stimulator for the bacterial LPS was found in this condition [70].
Pharmaceutical or genetic inhibition of the CB1 receptor cannabinoid protects against obesity, hepatic steatosis, and low-grade inflammation. In the context of monitoring the subsequent rat experiments, investigators concluded that the intestinal microbiome ECB behavior in the colon and adipose tissue (comparing the germ-free to standard mouse and analyzing the genetically engineered microbiome) was reported to be changed. The alleged role of the ECB system in healthy persons has yet to be known, but bacterial LPS can be induced and contributes to metabolic dysfunction [71].

3.2. Effects of Pre-/Probiotic Administration in MetS

While some clinical trials confirm the prediction that MetS is positively affected by probiotics and/or prebiotics, other experiments have shown contradictory findings, Table 1 summarizing them.
From its inception until 4 July 2019, in PubMed and Scopus, a qualitative systematic review was performed, according to the Cochrane approach, as well as a thorough literature search for randomized, controlled experiments. Nine clinical trials were finally reviewed according to our inclusion criteria, equal to six randomized control trials (RCTs).

3.2.1. Effects of Probiotics Alone

In some trials, the use of probiotics in patients with MetS has increased the index of body fat, BP, lipid profile, and glucose metabolism. In inflammatory biomarkers, probiotics also have a beneficial effect on molecule 1 (sVCAM-1), 6 (IL-6), α factor of necrosis (TNF-α), endothelial vascular factor of growth (VEGF), and thrombomodulin. The admission of probiotic compounds in patients with MetS leads to discreet changes in some clinical features of MetS and a decrease in inflammatory biomarkers, considering the diversity of the published reports [102].
In a Swedish analysis of the HFD-fed mice supplemented with L. plantarum, the lack of any reaction, specifically to the intravenous glucose-tolerance test, was related to the administration route that could not induce glucagon-like peptide-1 and release insulin. After collection from 426 strains, L. plantarum was used in a Korean sample as well [103].
The decline in insulin resistance parameters was associated to the cell reports of increased insulin signaling and inflammatory genes. L. casei interference improved the concentration of Lactobacillus and Bifidobacterium and decreased Clostridium in hyper insulinemic fructose-fed mice. The reduction of GLP-2 induced by Bacteroides fragilis was related to decreased insulinemia, while performing oral glucose tolerance tests (OGTT) [104]; moreover, some advantages were also found in liver function genes and gene expressions.
L. coryniformis administration with HFD-fed mice has caused vascular benefits and could be correlated with improvements in the composition of microbiota and intestinal permeability with reduced endotoxemia [105]. In research conducted in Taiwan, the use of lyophilized and “live” L. reuteri supplements increased Lactobacillus and lowered the abundance of pathogens, which helped improve the intestinal barrier. Weight gain, lipid profile change, hepatic steatosis, as well as inflammation and insulin resistance-related gene expressions decreased [106]. L. reuteri has also been used with the Aryl hydrocarbon receptor antimicrobial agonist MS in a French analysis. The addition improves glucose metabolism and liver alanine transaminase. Added synthesis increased glucose and hepatic alanine transaminase levels. In a Korean published paper, HFD-fed mice to whom L. sakei was administered as well, gained both weight and epididymal fat mass, by decreasing the inflammation and increasing the intestinal barrier (increased genetic expressions of tight-joint protein) [107]. Another two experiments in diabetic mice demonstrated similar decreases in insulin tolerance, TNF-α, and IL-6 at a dosage of 109 CFU pioglitazone and separate doses of L. casei [108]. The increased concentration of Lactobacillus and Bifidobacterium and SCFA has been linked to a potential rise in glucose homeostasis. Traditionally, probiotic activity stopped pancreatic islets from degeneration and/or regeneration.
An analysis comparing metformin, vildagliptin, and three L. rhamnosus strains (GG, MTCC5690, MTCC5689) increased the insulin tolerance curve areas and OGTT, with the exception of the HFD-MTCC5690 treatment [109]. Probiotics enhanced bowel permeability, increased adiponectin, and reduced inflammatory markers. Metabolic advantages gained through probiotics are similar to metformin and vildagliptin.
An Indian study on diabetic mice revealed greater results on the glucose metabolism, lipid profiles, and oxidation stress markers with L. rhamnosus NCDC17 compared to L. rhamnosus GG supplements. Moreover, adiponectin, pro-glucagon, and prohormone genes (TNF-α and IL-6) were found as being inflamed [110]. HFD-fed mice received Lactobacillus rhamnosus in order to minimize the abundance of Bilophila wadsworthy associated with MS [111]. Additionally, the B. wadsworthia increased in accordance with the effects of glucose metabolism, but the probiotic only enhanced the implications of HFD in the event that these bacteria were in high abundance.
In a Japanese study with L. gasseri in fermented milk, the complementation increased the production of insulin by reducing pancreatic/systemic inflammation [112]. L. paracasei was tested in a study for the advantages of glucose and lipid metabolism in obese rats and for many indicators of renal function and inflammation [113]. A research showing a decline in weight gain, visceral fat accumulation, and inflammation in combination with an increase in glucose and lipids studied the role of sterilized B. longum in obese mouse [114]. By comparing B. lactis and B. longum supplements, improved productivity of the former in terms of acetate and metabolic benefits was revealed [115]. For two doses of B. breve in HFD-induced mice, weight and visceral fat were decreased and genetic expressions decreased [116]. The authors also suggested that the probiotic could enhance healthy commensal bacteria to prevent endotoxemia and inflammation in an experiment with B. animalis [117]. The benefits of C. butyricum supplementation were complemented by increased intestinal and antimicrobial effects, supplementing glucose and lipid metabolism [118].

3.2.2. Effects of Associated Pre-/Probiotics and Synbiotics

Four studies have compared pro-/prebiotics and synbiotics (which are combination between pro- and prebiotics). Many of these have been xylo-oligosaccharide (XOS), inulin, and fructo-oligosaccharide and prebiotics (FOS). First, dysbiosis and inflammation were assessed in rats that were obese, with improved synbiotic results [119]. The groups treated also reduced the Firmicutes spp.-to-LPS ratio and raised the concentration of Bifidobacteria spp. in the prebiotic culture. The investigators concluded that prenatal, probiotic, and synbiotic products contribute to a decrease in endotoxemia and inflammation. The second research study investigated the link between cognitive function, L. paracasei, XOS, and the gut–brain axis in obese rats, which are shielded against insulin [120]. The glucose and lipid metabolism improved with prebiotics, probiotics, and synbiotic interventions, but the adiposity decreased with the XOS and synbiotic. Both products were beneficial, improving brain mitochondrial function and hippocampal plasticity, decreasing also the microglial activation. The third research study explored the association between psychiatric disorders and oxidative stress following addition or mixture of inulin or L. plantarum [121].
All therapy improves oxidative stress and psychoactive effects (depressive and anxiety-like behaviors). During the fourth study, in which HFD-fed mice were given L. paracasei, FOS, or their combination, the impacts on NAFLD were assessed [122]. Benefits during intraperitoneal GTT were considered as follows: increased gut barriers, decreasing LPS levels, and successful insulin signaling pathways were found in all of the treated populations. Steatosis improved, as well as dyslipidemia and inflammatory disorders.

3.2.3. Additional Results Obtained with Probiotics of Various Strains

In one study, Lactobacillus spp. and Bifidobacterium spp. were combined, and two other Lactobacillus spp. plus one Bifidobacterium spp. strains were added [123,124]; the first two mediated beneficial modifications in microbiota structure, bodily adiposity, insulin tolerance, and dyslipidemia in HFD-induced obese mouse. In comparison to the isolated effect of each probiotic on the composition, B. animalis had the most pronounced preventive effect on the structure [123]. Another Brazilian trial demonstrated the regulation of the composition of microbiota and increased intestinal permeability, endotoxemia, and inflammation in a mixture of three probiotics. The treatment also included a drop in insulin and leptin hypothalamic tolerance, which influenced the eating pattern (reduced food intake and weight gain) [124].

4. The Roles of A. muciniphila in the Onset of Metabolic Syndrome

The only Gram-negative emblematic Verrucomicrobia that is widespread in human intestinal mucosa is A. muciniphila [125]. Gene sequence analyses revealed that there are multiple gene candidates for mucin encoding and the single chromosome is contained in 2176 genes with a 55.8% GC content after the MucT type strain of A. muciniphila, as reported by Derrien (ATCC BAA-835 1/4 CIP107961T) [126,127]. This non-motile, oval-formed microorganism is purely anaerobic and with chemical organotrophic material that can withstand low levels of oxygen. A. muciniphila may create mucin-degrading enzymes and use mucin in the mucosal layer of the epithelium as a source of carbon and nitrogen. A. muciniphila splits these compounds into acetic and propionic compounds, releasing sulfate [128,129]. A. muciniphila also is 3 to 5% of the total gut microbiome population in healthy adult humans based on an analysis of its distinctive 16SrRNA signature, although this amount differs by several factors. In stable human beings, A. muciniphila has been closely linked to age. Its colonization starts at a young age and ranges between 5.0 and 8.8 log cells/g in a year equivalent to the adult stage but decreases in the elderly [130,131]. In comparison, in patients with metabolic disorders, A. muciniphila and a mucosal pathology varied, and the incidence of appendicitis and IBD was reversely associated with this [132]. In addition, a negative association of intestinal A. muciniphila with diabetes, obesity, and others MetS has been shown [133].
Dual control of A. muciniphila and metabolic disorders has demonstrated that both A. muciniphila excess and supplementation can have an effect on the host body. The anatomy of A. muciniphila can also be affected. The dissemination of A. muciniphila through early vancomycin therapy to the early intestinal colonization could help regulate the progression of autoimmune diabetes [134]. In an initial analysis, A. muciniphila was recognized as a potential new therapeutic agent for obese patients. Most studies have shown the advantages of using A. muciniphila in metabolic and obesity disorders for prevention and progress. T2DM is characterized by a lower A. muciniphila abundance, low inflammation, and an intestinal permeability disease [135]. The degree of A. muciniphila development can be used to determine the metabolic state of the body, for instance glucose homeostasis, serum lipids, and human adipocyte distribution. The reason for associating A. muciniphila with the production of obesity is not yet entirely explained. We have therefore analyzed the most recent studies into A. muciniphila’s function in obesity and learned more about its effects on the distinctive changes in the expression of pathways in metabolic homeostasis [136].

4.1. A. muciniphila and Obesity: Evidence from Mouse Models

All of the previous studies found a correlation between the caloric intake of A. muciniphila and its abundance. Prebiotic administration in high-fat diet mice eliminated the metabolic endotoxemia that characterizes the compromised MetS present among obese participants, decreased the overall fat mass, and decreased body weight [137]. These findings have in fact been strongly linked to an abundance of A. muciniphila [138]. The reduction of A. muciniphila and inflammatory markers were crucial in all circulatory parameters (i.e., glucose, insulin, leptin, and triglycerides), with 118 of the 13 genes implicated in the fatty oxidation, production, and oxidation of positive ties, whereas Bifidobacterium spp. were important. Three inflammatory features, leptin, and only two genes involved in the oxidation of fatty compounds are positively and negatively related.
Schneeberger et al. documented the inversely related levels of A. muciniphila in mice with inflammatory markers, lipid synthesis and insulin tolerance, cardiovascular risk, and adiposity markers in their plasma. After six consecutive weeks of HFD administration, the main effects were body weight gain and adiposity in mice. The results therefore suggest that an HFD particularly affects the gut bacteria and show that the abundance of A. muciniphila is decreasing steadily with sustained dietary care in mice. This bacterium also reduces, showing a causal impact, the disease development before the initiation of metabolic changes [139]. In comparison, the abundance of A. muciniphila increases and decreases in mice fed a diet rich in fish oil and lard, with greater regulation of the intestinal barrier system and less inflammatory tissue, which can be converted into germless receiving mice [55]. Other authors also found that the gut barrier dysfunction, weight, and fat gain of HFD-fed mice can be decreased at the same time with A. muciniphila [140,141]. The relation between age and A. muciniphila was finally identified in mice, as the intestinal level of the bacterium is lower in older mice. An HFD improves the adipose tissue and intestinal microbiological composition considerably compared to aging [139].

4.2. A. muciniphila and Obesity: Evidence from Human Studies

Emerging research assessed the correlation between A. muciniphila intestinal abundance and human body weight. There are proofs that these two variables have an opposite correlation [142,143]. The numbers of Bifidobacterium spp. and A. muciniphila and the number of Staphylococcus spp., Enterobacteriaceae spp., and Escherichia coli were found to decrease in overweight pregnancy. They were analyzed by quantitative real-time PCR for their gut microbiota composition. The increase in overall bacteria and Staphylococcus was linked in the whole population with increased plasma cholesterol levels, while the increased amount of Bacteroidites spp. was associated with an increased level of HDL and folic acid [144].
Twenty overweight or obese children and 20 average regular weight children aged 4–5 years were evaluated by Karlsson et. al. Interestingly, in obese/oversized infants, the A. muciniphila levels have decreased dramatically, whereas in the same class, the Gram-negative Enterobacteriaceae concentrations were significantly higher. Bifidobacterium levels in obese/overweight children were inversely linked to alanine aminotransferase (ALT) [145]. Since diabetes and overweight are linked to increased intestinal permeability and low inflammation, endotoxemia caused by LPS is considered as one of the causative agents of obesity-related metabolic disorders. An in vitro observation that confirms the epithelial barrier role of A. muciniphila may provide a working hypothesis to streamline in vivo proof that links decreased fecal A. muciniphila with diabetes and obesity rate. This could reveal a mechanism for protecting HFD obese mice from bacterial LPS endotoxemia. No relationship was currently assessed between A. muciniphila and hypothalamic food intake regulation markers. According to all data, it can be concluded that A. muciniphila influences the reaction of humans in terms of improving inflammation, insulin resistance, and glycemia on their diet of caloric constraint [146].

4.3. A. muciniphila Medicinal Role in the Treatment of Metabolic Conditions

The study of A. muciniphila has contributed to a better understanding of its possible therapeutic effects and actions in determining certain diseases and metabolic disorders. A. muciniphila, in particular, tends to be involved in the development of amine butyrate and in the propionate extracellular pool. It is also important in the development of sulfide hydrogen, which can be anti-inflammatory and a powerful antioxidant [147,148]. The involvement of A. muciniphila in metabolism enhances prebiotic intake. Both studies performed on animals with A. muciniphila revealed that it decreases body weight and fat mass rise, hepatic steatosis, inflammation, cholesterol, and atherosclerosis; it also enhances insulin sensitivity and restores intestinal barrier function by influencing various factors, such as the thickness of the mucosal membrane, close attachment proteins, antimicrobial peptides, and immunity. A. muciniphila works in particular on the immunomodulatory involvement of a special protein called Amuc 1100 [149].
Ottman et al. recently documented the increase of glucose resistance and a decline in body weight and fat gain in mice fed an HFD relative to the untreated mice with refined recombinant Amuc 1100 protein. The critical role of A. muciniphila in intestinal health, particularly in metabolic immunomodulation, is evident in this scenario. In order to increase their therapeutic application to gastrointestinal disease, future studies will also need to accentuate the main role of this microorganism and of its proteins in metabolic regulation and immune modulation [150].
The clinical studies focused on the obesity implying A. muciniphila and its associated biomarkers are presented in Table 2.
A summary of the beneficial effects of A. muciniphila treatment on MetS conditions is schematized in Figure 2.

5. The Interaction between Dietary Polyphenols (Prebiotics) with Gut Microbiota

Dietary polyphenols are considered naturally existing substances in plants or obtained from foods (i.e., cereals, coffee, fruit, tea, vegetables, and wine [167,168,169]. It is estimated that the small intestine consumes just 5–10% of the gross intake of polyphenol. Residual polyphenols, along with conjugates excreted into bile in the intestinal lumen, may accumulate in large intestinal lumps to a millimolar stage, where they are subjected to the enzymatic activity of the intestinal microbial group [170].
The gut microbiota and polyphenols have two major complementary relationships. On the other hand, after ingestion, dietary polyphenols are subject to a dynamic metabolism, associated with human and microbial enzymes, which contributes to the production of a broad variety of circulating and excreted metabolites and catabolic items. Polyphenols and their metabolites may on the contrary affect and cause a modification of the structure of gut microbiota via numerous interactions. Some studies support the possibility of modifying and producing variations in the populations of microflora by providing prebiotic effects and anti-pathogenic intestinal flora after dietary phenolic substances reach the gut microbiota, along with aromatic metabolites produced [165,166,167]. Human beings regard the biochemical transformation of dietary polyphenols as a significant concern. A minimal volume of dietary polyphenols is consumed by the small intestine, often after conjugation reactions such as de-glycosylation [166,170]. After absorption into the small bowel, polyphenolic compounds of lower complexity can undergo biotransformation in the enterocyte and then in the liver cell, by step I (oxidation, reduction and hydrolyses). These transformations produce a metabolite of water-soluble conjugates (glucuronide, sulfates, and methyl derivatives) that are rapidly released into the systemic circulation for subsequent organ drainage and urine excretion. The polyphenolic backbone of 90–95% unabsorbed polyphenols comprises colonic bacterial enzymes in the vast intestinal system that manufactures metabolites, which results in various physiological effects [171].
Colonic microflora could turn polyphenols into bioactive compounds that could affect the intestinal ecology and human health. The recommended concentrations will alter the composition of the gut microflora by inhibiting particular bacterial groups, as studies in humans and animal have revealed. Others will prosper in the open niche of the ecosystem [172].
Besides processing the derived food, the gut microbiota may perform a variety of bio-transformations of the polyphenols that enter the colon and influence their intake and bio disposability [172]. Specifically, some phenols in the metabolism of Clostridium spp. and Eubacterium spp. were reported to include isoflavone (daytzine), flavonol (quercetin and kaempferol), flavanone (naringenin and iso-xanthumol), and flavan-3 oil (catechin and epicatechin) [173]. Polyphenol colonic fermentation produces a wide range of biotransformation ingredients, but most of them are derived from phenyl, phenyl propionic, phenyl-butyric, valerical, valerolactone, phloroglucinol, and di-benzo-pyran urolithin A or urolithin B [174]. Microbiota degrade the parent polyphenolics in a variety of products, including phenolic acids such as 3-hydroxyphenyl acid, from flavanol routines synthesis (found in tomatoes, for example). The health benefits of these indirect and final materials arise with the assumption that certain bioavailability factors may be clarified for many biological effects traditionally linked to polyphenols (relative to that of the parent compounds) [175]. Phenolic acids can be found both in the plasma and urine after a meal, but some are activated by mammalian enzyme mechanisms (for example protein catabolism), which make it difficult to understand the plasma and urine levels of phenolic acid. A greater inhibition of anti-platelet aggregation has been shown by 3,4-dihydroxy (3,4DHPAA), one of the key intermedial metabolites of rutile-catabatic acid [176], as well as of secretion of monocytes of the proinflammatory cytokines TNF-a and IL-6 [177] than by the parent compound. It has also been demonstrated that phenolic acids suppress protein glycation [178]. However, most phenolic acid bioactivity research experiments are still based on in vitro models and more in vivo evidence is needed.
The absorption rate of polyphenol is very low and up to 90% of these substances linger in the colon [179]. They are metabolized through bacterial esterase, glucosidase, demethylation, dihydroxylation, and decarboxylation processes, resulting in smaller metabolites, several of which can be absorbed by the intestinal mucosa, such as phenolic acids and short-chain fatty acids. It is important to know that the microbial bioconversion potential of each organism influences and impacts their bioavailability on the final metabolites produced. In reality, since individuals have their own unique intestinal microbiota synthetic signature that can permit a fingerprint comparison, it can modulate the effect of polyphenol on host welfare by human intestinal microbiota composition [173]. On the other hand, the modulating microbiota of the intestinal ecosystem can be influenced by polyphenols and their metabolites. Certain phenolic compounds with bacteriostatic or bactericidal behavior were considered potential antimicrobial agents. In addition, they can also serve as inhibitors of infection-causing bacteria within intestinal and urinary tract cells, suggesting that such phenolic compounds are capable of being used as antimicrobial agents against human pathogens [180].

5.1. Polyphenol Transformation via Intestinal Microbiota

Polyphenols are not well absorbed by GI proximity and tissue responses to high-molecular PPs are now impaired by the metabolism of PP-rich food (such as tea, wine, chocolate, and fruit) [181]. Glycosidic bond cleavage and heterocyclic column breakdown, for example, are related to the biotransformation of polyphenols (PPs) (e.g., anthocyanins) via GM [182]. This biotransformation could be accomplished by different reactions, such as oxidation, hydroxylating, dehydrogenation, decarboxylation, isomerization, glycosylation, methylating, etc. Buddleoside flavonoids were transformed into aglycon acetins by human intestinal microflora, resulting in the transformation of metabolites into methylated and hydroxylated by-products. This research has defined the ability to transform flavonoids extensively, by just four bacterial species, such as Escherichia sp. 4, Escherichia sp. 34, Enterococcus sp. 45, and then Bacillum sp. 46 [183].
Maya-Apaza et al. suggested that the formation of large metabolites of 4-hydroxybenzol-propionic acids was the outcome of human GM formation during polyphenol fermentation (cyanidin-glycosyl-rutinoside, quercetin-rutinoside, chlorogenic, and neochlorogenic acids) [184]. The latest laboratory studies have shown that human genetic materials (caffeic acid, 3,4-dihydroxybenzoic acid, 2,4,6-tryhydroxybenzoic acid, coumarin, p-coumaric acid, ferulic acid, 2,4,6-trihydroxybenzaldehyde, and gallic acid) are produced by human GM fermentation in blackberry anthocyanin extract. The metabolites that were produced after 6 h of fermentation were caused by total cyanidin-3-glucoside degradation [185]. Recent research recorded the transformation of Lactobacillus acidophilus, genetically engineered microbes, into aglycons which can further be changed or used directly in dietary plant glycosides by other bacterial organisms [186].
It can be concluded that polyphenol degradation metabolites are predominantly responsible for the host’s health benefits based on the available evidence.

5.2. Modulatory Impact of Polyphenols on Intestinal Microbiota

A proper diet, which includes fruits and vegetables, leads to achieving the best health. Polyphenols (PPs), owing to the potent antioxidant role, have positive health effects, and are associated with fruit and vegetables [187]. The improvements in the bioavailability of PPs and their metabolites are also influencing the structure of the gut microbiota. Considering the data from in vitro studies, both the animal and human trials show, however, that selected PPs could serve as “prebiotics” and modify the ecology of the intestinal microbiota, thereby affecting host health. PPs also play a vital role in providing health outcomes, such as weight loss by GM change and host/microbe interactions [188]. A recent study by Sun et al. found that tea PPs obtained from the samples Bifidobacterium spp., Lactobacillus spp., and Enterococcus spp. have a significant impact on the gut microbiome [189]. The collections have increased SCFA efficacy and minimized the distribution of Prevotella, Bacteroides, and Clostridium histolyticum. In a study that shows the effect of black tea and red wine/grape extract on human gut microbiota, Kemperman et al. used the stimulated intestinal microbial environment, SHIME [190]. According to their study, the growth of Bifidobacteria, B. coccoides, Victivallis, and Anaeraeroglobus was improved by Klebsiella spp., Enterococci spp., and Akkermansia spp. simultaneously. The red wine/grape extract also encouraged the production of Alistipes spp., Cloacibacillus spp., Victivallis spp., and Akkermansia spp., and reduced the development of Bacterioidites spp., Bifidobacterium spp., B. coccoides, Subdoligranulum spp., and Anaeroblobus spp. It has been shown that red wine extract fermentation decreases the in vitro growing of Clostridium histolyticum, a harmful bacterial species in GM [191]. Similar findings were presented in a study carried out with human volunteers and in relation to Clostridium histolyticum development accompanied by red wine consumption [192]. On the other hand, Cueva and others note that the bacterial growth of the intestines can be greatly impaired by in vitro fermentation of the grape-derived flavan-3-ols [193].

5.3. Benefits of Polyphenols in MetS Patients

Many studies have shown that fruit-containing PPs play an important role in reducing the complications related to obesity [194,195,196]. An increasing part of the literature also points to a role of PPs in defending against diabetes and related complications by genetically modified organisms. The high prevalence of intestinal Bacteroidetes due to the improved potential for glycan degradation is caused by a PP-rich diet. Researchers thus hypothesized that microbiota modulation may contribute to the anti-obesity and antidiabetic mechanism through polyphenols [197]. The administration of polyphenol-rich cranberry extract (CE) avoided a high fructose–high sucrose dietary weight gain and visceral obesity development in mice. CE also decreased the production of triglycerides and prevented liver inflammation, thus improving the sensibility to insulin and avoiding the development of NAFLD. Furthermore, scientists observed that the positive outcomes of CE therapy are related to increasing Akkermansia’s abundance [198]. The effect of dietary supplements with grape polyphenols (i.e., a decline in the ratio of Firmicutes to Bacteroidetes) was substantially modulated in HFD-fed mice [199]. The GP supplementation has also stimulated A. muciniphila development and protected these improvements against the harmful effects of a high-fat diet [200]. The preventive impact against obesity and related risk factors has been identified with procyanidin supplement (PS) intake. PS treatment reduced weight gain, improved dyslipidemia, and reduced energy consumption. The positive influence of PS on obesity was also beneficial related to the effect of genetically modified modulation. The 16s gene sequencing RNA study showed that PS administration significantly increased Bacteroidetes β-diversity, intestinal microbiota, and the Firmicutes–Bacteroidetes ratio. Even an increase is considered to describe obesity-driven dysbiosis in Firmicutes/Bacteroidetes. In addition, studies suggest that an increased pathogenesis and the progression of obesity and T2DM have also been linked with Lachnospiraceae spp. [201].
Everard highlighted the opportunity to improve metabolic symptoms by supplementing with Akkermansia spp. as a probiotic for LPS prevention and intestinal permeability reduction [141]. Li and his colleagues found sinapine polyphenol to alter the intestinal microbiota in high-fat diet-induced mice in preventing NAFLD. It is a chronic low-grade inflammatory liver condition. The administration of Sinapine reduced the ratio of Firmicutes to Bacteroidetes and increased the abundance of Lactobacillaceae, Akkermansiaceae, and Blautia. Moreover, Sinapine stimulated G protein-coupled upregulation 43 (GPR43) of short-chain fatty acid (SCFA) in order to suppress the causes of inflammation [202,203,204]. A recent research study also found that PPs decreased microbiota dysbiosis by scavenging intestinal reactive oxygen species (ROS) through gut metabolism. Grape PPs suppressed the fat-driven generation of intestinal ROS and thereby decreased metabolic syndrome mediated by intestinal dysbiosis. Grape PPs stimulated healthy anaerobic intestinal bacteria, including A. muciniphila, which was linked with increased human metabolism [205]. These results revealed that dietary polyphenols may eradicate metabolic diseases by modulating intestinal microbiota positively.

5.4. Triggering A. muciniphila with Dietary Polyphenols

An increasing abundance of A. muciniphila, for instance, was correlated with antidiabetic effects of metformin in gut microbiota of obese rats in which obesity was induced through diet (DIO mice) [206,207].
A recent study has demonstrated that regular administration of polyphenol-rich cranberry extract (CE), orally, stopped weight gain over 8 weeks and enhanced many characteristics of metabolic syndrome, along with a major increase in Akkermansia’s abundance in DIO-mice intestinal microbiota [198]. That was the first confirmation that a fruit extract rich in polyphenol rich from fruits has a prebiotic effect on Akkermansia, which enhances the metabolic condition. These results have been supported by a more recent study on grape extracts, also being correlated with the prebiotic effect on Akkermansia, in increasing the metabolism in DIO mice [200]. In line with those observations, in addition to butter fat, the diet of mice was supplemented with California table grape powder, leading to lower adiposity and lipogenesis in conjunction with a higher trend in Akkermansia in intestinal microbiota [208]. Since both cranberries and grapes contain significant pro-anthocyanidin concentrations (PACs, also called condensed tannins), the unique class of polyphenols in this prebiotic operation can be considered of special significance. Cranberry PACs have also previously been correlated with increasing mucus production in mice and can provide Akkermansia with sufficient trophic resources [209].
Together with a propensity towards a growing degree of Akkermansia in gut microbiota, two new experiments have confirmed the benefits of administering flavonol-quercetin to obese rats [210]. Another study found that Akkermansia spp. was present in stool samples of healthy participants eating pomegranate extract (a rich source of ellagitannins) [211]; this new research being especially important, not just because it shows that human gut microbiota often has big effects on Akkermansia spp. when polyphenol rich extracts is called for, but also because healthy persons can benefit from the polyphenols that influence Akkermansia spp., which has prebiotic effects.
Interestingly enough, Roopchand et al. reported the colonic or jejunal expression of Muc2 mRNA after using Concord grape extract, type B PCs, and other polyphenols, which suggests that the Akkermansia niche has a direct effect. These findings, namely that polyphenols in grape juice and red wine increase Akkermansia spp.’s abundance, supported the hypothesis when applied to the in vitro intestine model [190].
Several clinical published data on Akkermansia spp. have shown an improvement in dietary obesity and are linked to a decline in weight gain, obesity, and increased glucose tolerance [206]. Live A. muciniphila administration has reversed obesity and metabolism by reducing adiposity, inflammation signs, insulin tolerance, and strengthening gut barrier in HFD mice [141]. Recently, the introduction of capsaicin (a dietary polyphenol) was shown to contribute to a reduction in weight gain, in mice fed with HFD, and excess of the Akkermansia spp., Bacteroides spp., and Coprococcus spp. [212]. Another work reveals that cranberry extract exerts positive metabolic effects by enhancing the dietary features caused by high fat/high sucrose (HFHS), which is linked to a proportional growth in Akkermansia spp. population [198].
The dietary replacement of an HFD with grape polyphenols contributed to dramatical improvements in the composition of the intestinal microbial population, which included a decline in the Firmicutes-to-Bacteroidetes ratio and an improvement in A. muciniphila. These modifications will provide some protection against the harmful effects of an HFD [200].

6. Pre-/Probiotic Formulations Using Nanotechnology: Applications in MetS

Emerging uses of nanotechnology [213] have recently been developed and applied in prebiotics and probiotics [214,215]. Probiotic nanotechnology is an evolving discipline that creates and allows entirely new options for the use of probiotics. In contrast with their use in medicines and pharmaceuticals, their uses in agriculture and the food industry are comparatively new. Currently, the production of nano-encapsulated probiotics is the fundamental application of probiotic nanotechnology. In order to improve flavor, texture, and consistency, the nanostructured food ingredients are created. Nanotechnology applications in producing organic food need vigilance, since their environmental and human health effects are not well established. There are currently no laws directly regulating or restricting the production of nanosized particles, primarily because of a lack of awareness of hazards [216].

6.1. Prebiotics Formulations

In the last few years, various forms of prebiotic formulations have gained further interest. The most favorable and most studied formulation of these prebiotics is the encapsulation into emulsions or structures dependent on nano and microparticles.
Maria et al. have therefore investigated the effect of the incorporation of numerous prebiotic fibers on the rheological and technological properties and the microstructure of an emulgated meat product (bologna). It was concluded that the stability of the released meat is improved with the inclusion of partial manhole starch and prebiotic fibers, to ensure that the provision of bologna sauce is safe [217].
Kazmierczak et al. identified a revolutionary strategy, loading these nanoparticles onto a biological carrier that delivers the treatments to a target site (a novel, nontoxic, therapeutic strain of Salmonella typhimurium that has been engineered as a biological delivery system hindering prostate cancer cells). Hu et al. also presented an example of nanoparticle-embodied probiotic bacteria that demonstrated an effective strategy to produce powerful and scalable DNA vaccines by covering live bacterial cells with synthetic nanoparticles [218,219]. Feher documented the use of nano-sized particles for prevention and treatment of neuroinflammation [220]. In reality, as an alternative to classical antibiotics to tackle bacterial resistance, probiotics attract the particular interest of people. Kim et al. recommended that prebiotics be produced to improve the function of probiotics by conjugating pathalic anhydride with dextran to create prebiotic formulations composed of Pediococcus acidilactidi [221]. They measured the cellular effects and the antimicrobial properties of the manufactured nanomaterial. Increased development of antimicrobial peptides using a self-defensing mechanism was observed for P. acidilactidi in phthalyl dextran nanoparticles, with an increased antimicrobial impact against both type of microorganisms—Gram (+) and Gram (−). Prebiotic phthalyl inulin nanoparticles were previously documented to boost P. acidilactici’s antimicrobial activities as well [222]. The increased antimicrobial activity of L. plantarum-treated phthalyl pullulan nanoparticles with Escherichia coli K99 and Listeria monocytogenes was also documented by Hong [223].

6.2. Probiotics Formulations

The nanoparticle was internalized by an energy-related and galactose-dependent conveyor mechanism in L. plantarum and higher amounts of plantaricin were secreted out from the nanoprobiotic produced as compared to the probiotic alone. The application of probiotic spores as a supply chain for chemotherapeutic drugs was recently proposed. In the autonomous production of nanoparticles in the gastrointestinal tract, Song et al. developed modified deoxycholic acid spores packed with doxorubicin and sorafenib [224]. Such a strategy targets the safety of medications in order to boost bioavailability in oral administration. In addition, disintegrated hydrophobic protein and hydrophilic deoxycholic acid are the source of the release, which increases the basolateral release of drugs by enhancing epithelial cells through the pathway to bile acid. Aziz identified the anticarcinogenic activity of nanoparticles of silver/Lactobacillus rhamnosus GG [225]. With the MTT test, the authors demonstrated that the viability of the HT-29 cell lines was substantially diminished, and apoptosis was caused by the addition of the maximum measured concentration of nanoparticles. It was observed that the synthesis of silver/Lactobacillus rhamnosus GG nanoparticles are cost efficient and offers a feasible biomedical nano-probiotic approach.
Fung’s work should be remembered, where the use of nanofibers to nanoencapsulate L. acidophilus, using 8 percent polyvinyl alcohol as a probiotic encapsulant for electrospinning technology, was suggested after investigating agro-waste-based nanofibers [226].
In Ebrahimnejad’s recent paper, chitosan is used as a probiotic bacteria to enhance the viability and survival of L. acidophilus nanoencapsulation towards gastrointestinal environments. Their usage as a probiotic bacterium was identified by the nanoencapsulation of probiotic bacteria [227].

6.3. Symbiotic Formulations

In a study of the impact of including inulin in alginate beads, Atia et al. developed an embedded oral-symbiotic supplement and observed its capacity to protect three different probiotic strains: P. acidilactici, L. reuteri, and L. salivarius. The bacteria were not found to be impaired by the encapsulation of the antimicrobial and probiotic properties [215].
A symbiotic nano-emulsion based on inulin was developed by Krithika and Preetha for enhancing probiotic stability; as a transport system for various probiotics in food products, the whey protein concentrate/inulin nano complex may be proposed [228]. Salmeron et al. published their work on the production of the fermented beverages with synbiotic properties in the field of food produced to boost human health and well-being by integrating specific and special bioactive nanoparticles, to enhance organoleptic proprieties, nutrient absorption, and the supply of bioactive nutrients and compounds [229].

7. Concluding Remarks and Future Perspectives

The administration of prebiotics and/or probiotics in MS is accompanied by numerous benefits in terms of improving metabolic parameters such as BMI, abdominal circumference, basal or postprandial glycemia, insulin resistance, and inflammation parameters. The quality of the microbial flora also influences numerous pathophysiological processes specific to MetS and T2DM (i.e., insulin resistance, pro-inflammatory status, regulation of blood sugar or appetite, etc.). In recent years, the attention paid to the bacterium A. muciniphilia is due to the observed and proven benefits in reducing body weight, with numerous studies (as highlighted in this review) supporting the need for colonization with Akkermansia spp. Moreover, the consumption of foods rich in polyphenols positively influences the bacterial microflora. Nanotechnology can also offer numerous solutions for overcoming the gastric barrier, as well as for achieving adequate concentrations of compounds at the intestinal level, the administration of pre-/probiotics being one of the applications of this expanding field.
Overall, several studies have shown that favorable manipulation of the probiostatic or prebiotic usage of the intestinal microbiota and the immune system will boost typical parameters of MetS. However, more properly planned animal and human experiments may reveal new information in order to justify the controversies surrounding the impact and doses of pre-/probiotics on the MetS risk factors and offer a more complete explanation of the function of the acts concerned. Specific clarified considerations include the minimum required for producing beneficial benefits, the duration of supplementation, persistence of this influence, and potential contraindications. Diverse hypotheses were newly introduced to clarify the advantages of probiosis to methyl microorganisms; however, there were substantial issues that must be considered with respect to probiotic effects and their action mechanisms regarding the strain specificities and duration of administration. Furthermore, the chemical composition of prebiotic compounds is a crucial consideration for the regulation of microorganisms of the intestinal microbiota. To explain the efficacy of pre-/probiotics on MetS prevention and management and endorse their future application of clinical procedures, scientifically randomized, placebo-controlled trials, utilizing, for example, multiple subjects, should be undertaken [230].
Pharmacists are encouraged to be familiar with pre-/probiotic products that are generally available so that they can make reasonable decisions for particular groups of patients. It is important for these drug specialists to realize when a probiotic should be prescribed, that not all strains are the same, and that certain benefits of this type of supplement are unique to each type of condition or disorder. Equipped with proper expertise, pharmacists may advise patients related to the strains or mixtures, in close correlation with the action/therapeutic advantage that the subject is looking for.
Of course, there are some additional features of a quality probiotic product, as well as a scientifically sustained benefit that must be mentioned. The following data should always be included on the probiotic label: adequate storage directions; CFU amount before date of expiry; date of expiry; details on the producer company’s contact; microbe names/genus/species/strains; and the prescribed dosage or section in compliance with clinical evidence, etc. When a probiotic is recommended, it must be well-known that not every strain is the same, that the benefits are strain-specific, so the patient must pick, related to the advantage, a scientifically proven strain. Moreover, they must select a probiotic according to scientific data, at the dosage given for that strain (or mixture of strains), and be careful when considering the following specific aspects: a high CFU count (more not being always good) and probiotic formulations of multi strains (many of them may not having clinical evidence).

Author Contributions

All authors have equal contribution to it and have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

The authors wish to thank to University of Oradea for its technical support in conducting this research.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hays, N.P.; Galassetti, P.R.; Coker, R.H. Prevention and treatment of type 2 diabetes: Current role of lifestyle, natural product, and pharmacological interventions. Pharmacol. Ther. 2008, 118, 181–191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Monteagudo-Mera, A.; Rastall, R.A.; Gibson, G.R.; Charalampopoulos, D.; Chatzifragkou, A. Adhesion mechanisms mediated by probiotics and prebiotics and their potential impact on human health. Appl. Microbiol. Biotechnol. 2019, 103, 6463–6472. [Google Scholar] [CrossRef] [Green Version]
  3. Pandey, K.R.; Naik, S.R.; Vakil, B.V. Probiotics, prebiotics and synbiotics—A review. J. Food Sci. Technol. 2015, 52, 7577–7587. [Google Scholar] [CrossRef]
  4. Gibson, G.R.; Roberfroid, M.B. Dietary modulation of the human colonic microbiota: Introducing the concept of prebiotics. J. Nutr. 1995, 125, 1401–1412. [Google Scholar] [CrossRef] [PubMed]
  5. Kneifel, W. In vitro growth behaviour of probiotic bacteria in culture media with carbohydrates of prebiotic importance. Microb. Ecol. Health Dis. 2000, 12, 27–34. [Google Scholar] [CrossRef]
  6. Cheng, K.C.; Demirci, A.; Catchmark, J.M. Pullulan: Biosynthesis, production, and applications. Appl. Microbiol. Biotechnol. 2011, 92, 29–44. [Google Scholar] [CrossRef]
  7. Catley, B.J.; Ramsay, A.; Servis, C. Observations on the structure of the fungal extracellular polysaccharide, pullulan. Carbohydr. Res. 1986, 153, 79–86. [Google Scholar] [CrossRef]
  8. Leathers, T.D. Biotechnological production and applications of pullulan. Appl. Microbiol. Biotechnol. 2003, 62, 468–473. [Google Scholar] [CrossRef]
  9. Sugawa-Katayama, Y.; Kondou, F.; Mandai, T.; Yoneyama, M. Effects of Pullulan, Polydextrose and Pectin on Cecal Microflora. J. Appl. Glycosci. 1994, 41, 413–418. [Google Scholar] [CrossRef]
  10. Pineiro, M.; Asp, N.G.; Reid, G.; Macfarlane, S.; Morelli, L.; Brunser, O.; Tuohy, K. FAO Technical meeting on prebiotics. J. Clin. Gastroenterol. 2008, 42 (Suppl. 3), S156–S159. [Google Scholar] [CrossRef]
  11. Mohamadshahi, M.; Veissi, M.; Haidari, F.; Javid, A.Z.; Mohammadi, F.; Shirbeigi, E. Effects of probiotic yogurt consumption on lipid profile in type 2 diabetic patients: A randomized controlled clinical trial. J. Res. Med. Sci. 2014, 19, 531–536. [Google Scholar] [PubMed]
  12. Parker, R.B. Probiotics, the other half of the antibiotic story. Anim. Nutr. Health 1974, 29, 4–8. [Google Scholar]
  13. Havenaar, R.; Huis In’t Veld, J.H.J. Probiotics: A General View. In The Lactic Acid Bacteria Volume 1: The Lactic Acid Bacteria in Health and Disease; Wood, B.J.B., Ed.; Springer: Boston, MA, USA, 1992; pp. 151–170. [Google Scholar] [CrossRef]
  14. Sanders, M.E.; Merenstein, D.J.; Reid, G.; Gibson, G.R.; Rastall, R.A. Probiotics and prebiotics in intestinal health and disease: From biology to the clinic. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 605–616. [Google Scholar] [CrossRef]
  15. Reid, G.; Sanders, M.E.; Gaskins, H.R.; Gibson, G.R.; Mercenier, A.; Rastall, R.; Roberfroid, M.; Rowland, I.; Cherbut, C.; Klaenhammer, T.R. New scientific paradigms for probiotics and prebiotics. J. Clin. Gastroenterol. 2003, 37, 105–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Sanz, Y.; Nadal, I.; Sánchez, E. Probiotics as drugs against human gastrointestinal infections. Recent Pat. Anti-Infect Drug Discov. 2007, 2, 148–156. [Google Scholar] [CrossRef]
  17. Furet, J.P.; Kong, L.C.; Tap, J.; Poitou, C.; Basdevant, A.; Bouillot, J.L.; Mariat, D.; Corthier, G.; Doré, J.; Henegar, C.; et al. Differential adaptation of human gut microbiota to bariatric surgery-induced weight loss: Links with metabolic and low-grade inflammation markers. Diabetes 2010, 59, 3049–3057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Cani, P.D.; Bibiloni, R.; Knauf, C.; Waget, A.; Neyrinck, A.M.; Delzenne, N.M.; Burcelin, R. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes 2008, 57, 1470–1481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Vallianou, N.G.; Stratigou, T.; Tsagarakis, S. Microbiome and diabetes: Where are we now? Diabetes Res. Clin. Pract. 2018, 146, 111–118. [Google Scholar] [CrossRef] [PubMed]
  20. Festi, D.; Schiumerini, R.; Eusebi, L.H.; Marasco, G.; Taddia, M.; Colecchia, A. Gut microbiota and metabolic syndrome. World J. Gastroenterol. 2014, 20, 16079–16094. [Google Scholar] [CrossRef]
  21. Shang, Q.; Song, G.; Zhang, M.; Shi, J.; Xu, C.; Hao, J.; Li, G.; Yu, G. Dietary fucoidan improves metabolic syndrome in association with increased Akkermansia population in the gut microbiota of high-fat diet-fed mice. J. Funct. Foods 2017, 28, 138–146. [Google Scholar] [CrossRef]
  22. Zhou, K. Strategies to promote abundance of Akkermansia muciniphila, an emerging probiotics in the gut, evidence from dietary intervention studies. J. Funct. Foods 2017, 33, 194–201. [Google Scholar] [CrossRef]
  23. Ley, R.E.; Bäckhed, F.; Turnbaugh, P.; Lozupone, C.A.; Knight, R.D.; Gordon, J.I. Obesity alters gut microbial ecology. Proc. Natl. Acad. Sci. USA 2005, 102, 11070–11075. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Roberfroid, M.; Gibson, G.R.; Hoyles, L.; McCartney, A.L.; Rastall, R.; Rowland, I.; Wolvers, D.; Watzl, B.; Szajewska, H.; Stahl, B.; et al. Prebiotic effects: Metabolic and health benefits. Br. J. Nutr. 2010, 104 (Suppl. 2), S1–S63. [Google Scholar] [CrossRef] [Green Version]
  25. Mozes, S.; Bujnáková, D.; Sefcíková, Z.; Kmet, V. Developmental changes of gut microflora and enzyme activity in rat pups exposed to fat-rich diet. Obesity 2008, 16, 2610–2615. [Google Scholar] [CrossRef]
  26. Mozes, S.; Bujnáková, D.; Sefcíková, Z.; Kmet, V. Intestinal microflora and obesity in rats. Folia Microbiol. 2008, 53, 225–228. [Google Scholar] [CrossRef] [PubMed]
  27. Turnbaugh, P.J.; Bäckhed, F.; Fulton, L.; Gordon, J.I. Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host Microbe 2008, 3, 213–223. [Google Scholar] [CrossRef] [Green Version]
  28. Turnbaugh, P.J.; Hamady, M.; Yatsunenko, T.; Cantarel, B.L.; Duncan, A.; Ley, R.E.; Sogin, M.L.; Jones, W.J.; Roe, B.A.; Affourtit, J.P.; et al. A core gut microbiome in obese and lean twins. Nature 2009, 457, 480–484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Le Chatelier, E.; Nielsen, T.; Qin, J.; Prifti, E.; Hildebrand, F.; Falony, G.; Almeida, M.; Arumugam, M.; Batto, J.M.; Kennedy, S.; et al. Richness of human gut microbiome correlates with metabolic markers. Nature 2013, 500, 541–546. [Google Scholar] [CrossRef]
  30. Plovier, H.; Cani, P.D. Microbial Impact on Host Metabolism: Opportunities for Novel Treatments of Nutritional Disorders? Microbiol. Spectr. 2017, 5. [Google Scholar] [CrossRef]
  31. Ussar, S.; Griffin, N.W.; Bezy, O.; Fujisaka, S.; Vienberg, S.; Softic, S.; Deng, L.; Bry, L.; Gordon, J.I.; Kahn, C.R. Interactions between Gut Microbiota, Host Genetics and Diet Modulate the Predisposition to Obesity and Metabolic Syndrome. Cell Metab. 2015, 22, 516–530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. de Cossío, L.F.; Fourrier, C.; Sauvant, J.; Everard, A.; Capuron, L.; Cani, P.D.; Layé, S.; Castanon, N. Impact of prebiotics on metabolic and behavioral alterations in a mouse model of metabolic syndrome. Brain Behav. Immun. 2017, 64, 33–49. [Google Scholar] [CrossRef] [PubMed]
  33. Li, T.; Gao, J.; Du, M.; Mao, X. Milk fat globule membrane supplementation modulates the gut microbiota and attenuates metabolic endotoxemia in high-fat diet-fed mice. J. Funct. Foods 2018, 47, 56–65. [Google Scholar] [CrossRef]
  34. Boutagy, N.E.; McMillan, R.P.; Frisard, M.I.; Hulver, M.W. Metabolic endotoxemia with obesity: Is it real and is it relevant? Biochimie 2016, 124, 11–20. [Google Scholar] [CrossRef] [Green Version]
  35. Frazier, T.H.; DiBaise, J.K.; McClain, C.J. Gut microbiota, intestinal permeability, obesity-induced inflammation, and liver injury. JPEN J. Parenter. Enter. Nutr. 2011, 35, 14s–20s. [Google Scholar] [CrossRef] [PubMed]
  36. Frank, D.N.; Zhu, W.; Sartor, R.B.; Li, E. Investigating the biological and clinical significance of human dysbioses. Trends Microbiol. 2011, 19, 427–434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Norris, G.H.; Jiang, C.; Ryan, J.; Porter, C.M.; Blesso, C.N. Milk sphingomyelin improves lipid metabolism and alters gut microbiota in high fat diet-fed mice. J. Nutr. Biochem. 2016, 30, 93–101. [Google Scholar] [CrossRef]
  38. Breban, M. Gut microbiota and inflammatory joint diseases. Jt. Bone Spine 2016, 83, 645–649. [Google Scholar] [CrossRef]
  39. Tejada-Simon, M.V.; Lee, J.H.; Ustunol, Z.; Pestka, J.J. Ingestion of yogurt containing Lactobacillus acidophilus and Bifidobacterium to potentiate immunoglobulin A responses to cholera toxin in mice. J. Dairy Sci. 1999, 82, 649–660. [Google Scholar] [CrossRef]
  40. Schertzer, J.D.; Tamrakar, A.K.; Magalhães, J.G.; Pereira, S.; Bilan, P.J.; Fullerton, M.D.; Liu, Z.; Steinberg, G.R.; Giacca, A.; Philpott, D.J.; et al. NOD1 activators link innate immunity to insulin resistance. Diabetes 2011, 60, 2206–2215. [Google Scholar] [CrossRef]
  41. Delzenne, N.M.; Neyrinck, A.M.; Cani, P.D. Modulation of the gut microbiota by nutrients with prebiotic properties: Consequences for host health in the context of obesity and metabolic syndrome. Microb. Cell Fact. 2011, 10, S10. [Google Scholar] [CrossRef] [Green Version]
  42. Awoyemi, A.; Trøseid, M.; Arnesen, H.; Solheim, S.; Seljeflot, I. Markers of metabolic endotoxemia as related to metabolic syndrome in an elderly male population at high cardiovascular risk: A cross-sectional study. Diabetol. Metab. Syndr. 2018, 10, 59. [Google Scholar] [CrossRef] [Green Version]
  43. Kang, Y.B.; Cai, Y.; Zhang, H. Gut microbiota and allergy/asthma: From pathogenesis to new therapeutic strategies. Allergol. Immunopathol. 2017, 45, 305–309. [Google Scholar] [CrossRef]
  44. Qian, L.L.; Li, H.T.; Zhang, L.; Fang, Q.C.; Jia, W.P. Effect of the Gut Microbiota on Obesity and Its Underlying Mechanisms: An Update. Biomed. Environ. Sci. 2015, 28, 839–847. [Google Scholar] [CrossRef]
  45. Moran, C.P.; Shanahan, F. Gut microbiota and obesity: Role in aetiology and potential therapeutic target. Best Pract. Res. Clin. Gastroenterol. 2014, 28, 585–597. [Google Scholar] [CrossRef] [PubMed]
  46. Hasnain, S.Z.; Borg, D.J.; Harcourt, B.E.; Tong, H.; Sheng, Y.H.; Ng, C.P.; Das, I.; Wang, R.; Chen, A.C.; Loudovaris, T.; et al. Glycemic control in diabetes is restored by therapeutic manipulation of cytokines that regulate beta cell stress. Nat. Med. 2014, 20, 1417–1426. [Google Scholar] [CrossRef]
  47. Burcelin, R.; Garidou, L.; Pomié, C. Immuno-microbiota cross and talk: The new paradigm of metabolic diseases. Semin. Immunol. 2012, 24, 67–74. [Google Scholar] [CrossRef] [PubMed]
  48. David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–563. [Google Scholar] [CrossRef] [Green Version]
  49. Turnbaugh, P.J.; Ley, R.E.; Mahowald, M.A.; Magrini, V.; Mardis, E.R.; Gordon, J.I. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006, 444, 1027–1031. [Google Scholar] [CrossRef]
  50. Devkota, S.; Wang, Y.; Musch, M.W.; Leone, V.; Fehlner-Peach, H.; Nadimpalli, A.; Antonopoulos, D.A.; Jabri, B.; Chang, E.B. Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il10−/− mice. Nature 2012, 487, 104–108. [Google Scholar] [CrossRef] [Green Version]
  51. Wan, Y.; Wang, F.; Yuan, J.; Li, J.; Jiang, D.; Zhang, J.; Li, H.; Wang, R.; Tang, J.; Huang, T.; et al. Effects of dietary fat on gut microbiota and faecal metabolites, and their relationship with cardiometabolic risk factors: A 6-month randomised controlled-feeding trial. Gut 2019, 68, 1417–1429. [Google Scholar] [CrossRef] [Green Version]
  52. Sonnenburg, E.D.; Smits, S.A.; Tikhonov, M.; Higginbottom, S.K.; Wingreen, N.S.; Sonnenburg, J.L. Diet-induced extinctions in the gut microbiota compound over generations. Nature 2016, 529, 212–215. [Google Scholar] [CrossRef] [Green Version]
  53. Desai, M.S.; Seekatz, A.M.; Koropatkin, N.M.; Kamada, N.; Hickey, C.A.; Wolter, M.; Pudlo, N.A.; Kitamoto, S.; Terrapon, N.; Muller, A.; et al. A Dietary Fiber-Deprived Gut Microbiota Degrades the Colonic Mucus Barrier and Enhances Pathogen Susceptibility. Cell 2016, 167, 1339–1353.e21. [Google Scholar] [CrossRef] [Green Version]
  54. Desbois, A.P.; Smith, V.J. Antibacterial free fatty acids: Activities, mechanisms of action and biotechnological potential. Appl. Microbiol. Biotechnol. 2010, 85, 1629–1642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Caesar, R.; Tremaroli, V.; Kovatcheva-Datchary, P.; Cani, P.D.; Bäckhed, F. Crosstalk between Gut Microbiota and Dietary Lipids Aggravates WAT Inflammation through TLR Signaling. Cell Metab. 2015, 22, 658–668. [Google Scholar] [CrossRef] [Green Version]
  56. Zhang, X.; Shen, D.; Fang, Z.; Jie, Z.; Qiu, X.; Zhang, C.; Chen, Y.; Ji, L. Human gut microbiota changes reveal the progression of glucose intolerance. PLoS ONE 2013, 8, e71108. [Google Scholar] [CrossRef]
  57. Yassour, M.; Lim, M.Y.; Yun, H.S.; Tickle, T.L.; Sung, J.; Song, Y.-M.; Lee, K.; Franzosa, E.A.; Morgan, X.C.; Gevers, D.; et al. Sub-clinical detection of gut microbial biomarkers of obesity and type 2 diabetes. Genome Med. 2016, 8, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Li, J.; Zhao, F.; Wang, Y.; Chen, J.; Tao, J.; Tian, G.; Wu, S.; Liu, W.; Cui, Q.; Geng, B.; et al. Gut microbiota dysbiosis contributes to the development of hypertension. Microbiome 2017, 5, 14. [Google Scholar] [CrossRef] [Green Version]
  59. Depommier, C.; Everard, A.; Druart, C.; Plovier, H.; Van Hul, M.; Vieira-Silva, S.; Falony, G.; Raes, J.; Maiter, D.; Delzenne, N.M.; et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: A proof-of-concept exploratory study. Nat. Med. 2019, 25, 1096–1103. [Google Scholar] [CrossRef] [PubMed]
  60. Wang, Z.; Klipfell, E.; Bennett, B.J.; Koeth, R.; Levison, B.S.; Dugar, B.; Feldstein, A.E.; Britt, E.B.; Fu, X.; Chung, Y.M.; et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 2011, 472, 57–63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Tang, W.H.; Wang, Z.; Levison, B.S.; Koeth, R.A.; Britt, E.B.; Fu, X.; Wu, Y.; Hazen, S.L. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N. Engl. J. Med. 2013, 368, 1575–1584. [Google Scholar] [CrossRef] [Green Version]
  62. Zhu, W.; Gregory, J.C.; Org, E.; Buffa, J.A.; Gupta, N.; Wang, Z.; Li, L.; Fu, X.; Wu, Y.; Mehrabian, M.; et al. Gut Microbial Metabolite TMAO Enhances Platelet Hyperreactivity and Thrombosis Risk. Cell 2016, 165, 111–124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Larsen, G.L.; Henson, P.M. Mediators of inflammation. Annu. Rev. Immunol. 1983, 1, 335–359. [Google Scholar] [CrossRef] [PubMed]
  64. Hotamisligil, G.S.; Shargill, N.S.; Spiegelman, B.M. Adipose expression of tumor necrosis factor-alpha: Direct role in obesity-linked insulin resistance. Science 1993, 259, 87–91. [Google Scholar] [CrossRef] [PubMed]
  65. Hotamisligil, G.S.; Arner, P.; Caro, J.F.; Atkinson, R.L.; Spiegelman, B.M. Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance. J. Clin. Investig. 1995, 95, 2409–2415. [Google Scholar] [CrossRef] [PubMed]
  66. Hotamisligil, G.S. Inflammation, metaflammation and immunometabolic disorders. Nature 2017, 542, 177–185. [Google Scholar] [CrossRef] [PubMed]
  67. O’Rourke, R.W.; White, A.E.; Metcalf, M.D.; Olivas, A.S.; Mitra, P.; Larison, W.G.; Cheang, E.C.; Varlamov, O.; Corless, C.L.; Roberts, C.T., Jr.; et al. Hypoxia-induced inflammatory cytokine secretion in human adipose tissue stromovascular cells. Diabetologia 2011, 54, 1480–1490. [Google Scholar] [CrossRef] [Green Version]
  68. Weisberg, S.P.; McCann, D.; Desai, M.; Rosenbaum, M.; Leibel, R.L.; Ferrante, A.W., Jr. Obesity is associated with macrophage accumulation in adipose tissue. J. Clin. Investig. 2003, 112, 1796–1808. [Google Scholar] [CrossRef] [PubMed]
  69. Byndloss, M.X.; Olsan, E.E.; Rivera-Chávez, F.; Tiffany, C.R.; Cevallos, S.A.; Lokken, K.L.; Torres, T.P.; Byndloss, A.J.; Faber, F.; Gao, Y.; et al. Microbiota-activated PPAR-γ signaling inhibits dysbiotic Enterobacteriaceae expansion. Science 2017, 357, 570–575. [Google Scholar] [CrossRef]
  70. Muccioli, G.G.; Naslain, D.; Bäckhed, F.; Reigstad, C.S.; Lambert, D.M.; Delzenne, N.M.; Cani, P.D. The endocannabinoid system links gut microbiota to adipogenesis. Mol. Syst. Biol. 2010, 6, 392. [Google Scholar] [CrossRef] [PubMed]
  71. Geurts, L.; Lazarevic, V.; Derrien, M.; Everard, A.; Van Roye, M.; Knauf, C.; Valet, P.; Girard, M.; Muccioli, G.G.; François, P.; et al. Altered gut microbiota and endocannabinoid system tone in obese and diabetic leptin-resistant mice: Impact on apelin regulation in adipose tissue. Front. Microbiol. 2011, 2, 149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Guardamagna, O.; Amaretti, A.; Puddu, P.E.; Raimondi, S.; Abello, F.; Cagliero, P.; Rossi, M. Bifidobacteria supplementation: Effects on plasma lipid profiles in dyslipidemic children. Nutrition 2014, 30, 831–836. [Google Scholar] [CrossRef]
  73. Ooi, L.G.; Ahmad, R.; Yuen, K.H.; Liong, M.T. Lactobacillus acidophilus CHO-220 and inulin reduced plasma total cholesterol and low-density lipoprotein cholesterol via alteration of lipid transporters. J. Dairy Sci. 2010, 93, 5048–5058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Ryan, J.J.; Hanes, D.A.; Schafer, M.B.; Mikolai, J.; Zwickey, H. Effect of the Probiotic Saccharomyces boulardii on Cholesterol and Lipoprotein Particles in Hypercholesterolemic Adults: A Single-Arm, Open-Label Pilot Study. J. Altern. Complement. Med. 2015, 21, 288–293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Xu, J.; Lazou Ahrén, I.; Olsson, C.; Jeppsson, B.; Ahrné, S.; Molin, G. Oral and faecal microbiota in volunteers with hypertension in a double blind, randomised placebo controlled trial with probiotics and fermented bilberries. J. Funct. Foods 2015, 18, 275–288. [Google Scholar] [CrossRef]
  76. Hata, Y.; Yamamoto, M.; Ohni, M.; Nakajima, K.; Nakamura, Y.; Takano, T. A placebo-controlled study of the effect of sour milk on blood pressure in hypertensive subjects. Am. J. Clin. Nutr. 1996, 64, 767–771. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Agerholm-Larsen, L.; Raben, A.; Haulrik, N.; Hansen, A.S.; Manders, M.; Astrup, A. Effect of 8 week intake of probiotic milk products on risk factors for cardiovascular diseases. Eur. J. Clin. Nutr. 2000, 54, 288–297. [Google Scholar] [CrossRef]
  78. Jones, M.L.; Martoni, C.J.; Di Pietro, E.; Simon, R.R.; Prakash, S. Evaluation of clinical safety and tolerance of a Lactobacillus reuteri NCIMB 30242 supplement capsule: A randomized control trial. Regul. Toxicol. Pharm. 2012, 63, 313–320. [Google Scholar] [CrossRef]
  79. Sharafedtinov, K.K.; Plotnikova, O.A.; Alexeeva, R.I.; Sentsova, T.B.; Songisepp, E.; Stsepetova, J.; Smidt, I.; Mikelsaar, M. Hypocaloric diet supplemented with probiotic cheese improves body mass index and blood pressure indices of obese hypertensive patients—A randomized double-blind placebo-controlled pilot study. Nutr. J. 2013, 12, 138. [Google Scholar] [CrossRef] [Green Version]
  80. Sperry, M.F.; Silva, H.L.A.; Balthazar, C.F.; Esmerino, E.A.; Verruck, S.; Prudencio, E.S.; Neto, R.P.C.; Tavares, M.I.B.; Peixoto, J.C.; Nazzaro, F.; et al. Probiotic Minas Frescal cheese added with L. casei 01: Physicochemical and bioactivity characterization and effects on hematological/biochemical parameters of hypertensive overweighted women—A randomized double-blind pilot trial. J. Funct. Foods 2018, 45, 435–443. [Google Scholar] [CrossRef]
  81. Mohammadi-Sartang, M.; Bellissimo, N.; Totosy de Zepetnek, J.O.; Brett, N.R.; Mazloomi, S.M.; Fararouie, M.; Bedeltavana, A.; Famouri, M.; Mazloom, Z. The effect of daily fortified yogurt consumption on weight loss in adults with metabolic syndrome: A 10-week randomized controlled trial. Nutr. Metab. Cardiovasc. Dis. 2018, 28, 565–574. [Google Scholar] [CrossRef]
  82. Kassaian, N.; Feizi, A.; Aminorroaya, A.; Amini, M. Probiotic and synbiotic supplementation could improve metabolic syndrome in prediabetic adults: A randomized controlled trial. Diabetes Metab. Syndr. 2019, 13, 2991–2996. [Google Scholar] [CrossRef] [PubMed]
  83. Rezazadeh, L.; Gargari, B.P.; Jafarabadi, M.A.; Alipour, B. Effects of probiotic yogurt on glycemic indexes and endothelial dysfunction markers in patients with metabolic syndrome. Nutrition 2019, 62, 162–168. [Google Scholar] [CrossRef]
  84. Leber, B.; Tripolt, N.J.; Blattl, D.; Eder, M.; Wascher, T.C.; Pieber, T.R.; Stauber, R.; Sourij, H.; Oettl, K.; Stadlbauer, V. The influence of probiotic supplementation on gut permeability in patients with metabolic syndrome: An open label, randomized pilot study. Eur. J. Clin. Nutr. 2012, 66, 1110–1115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Tripolt, N.J.; Leber, B.; Blattl, D.; Eder, M.; Wonisch, W.; Scharnagl, H.; Stojakovic, T.; Obermayer-Pietsch, B.; Wascher, T.C.; Pieber, T.R.; et al. Short communication: Effect of supplementation with Lactobacillus casei Shirota on insulin sensitivity, β-cell function, and markers of endothelial function and inflammation in subjects with metabolic syndrome—A pilot study. J. Dairy Sci. 2013, 96, 89–95. [Google Scholar] [CrossRef]
  86. Barreto, F.M.; Colado Simão, A.N.; Morimoto, H.K.; Batisti Lozovoy, M.A.; Dichi, I.; Helena da Silva Miglioranza, L. Beneficial effects of Lactobacillus plantarum on glycemia and homocysteine levels in postmenopausal women with metabolic syndrome. Nutrition 2014, 30, 939–942. [Google Scholar] [CrossRef] [PubMed]
  87. Bernini, L.J.; Simão, A.N.; Alfieri, D.F.; Lozovoy, M.A.; Mari, N.L.; de Souza, C.H.; Dichi, I.; Costa, G.N. Beneficial effects of Bifidobacterium lactis on lipid profile and cytokines in patients with metabolic syndrome: A randomized trial. Effects of probiotics on metabolic syndrome. Nutrition 2016, 32, 716–719. [Google Scholar] [CrossRef] [PubMed]
  88. Genta, S.; Cabrera, W.; Habib, N.; Pons, J.; Carillo, I.M.; Grau, A.; Sánchez, S. Yacon syrup: Beneficial effects on obesity and insulin resistance in humans. Clin. Nutr. 2009, 28, 182–187. [Google Scholar] [CrossRef] [PubMed]
  89. Kim, J.; Yun, J.M.; Kim, M.K.; Kwon, O.; Cho, B. Lactobacillus gasseri BNR17 Supplementation Reduces the Visceral Fat Accumulation and Waist Circumference in Obese Adults: A Randomized, Double-Blind, Placebo-Controlled Trial. J. Med. Food 2018, 21, 454–461. [Google Scholar] [CrossRef]
  90. Ivey, K.L.; Hodgson, J.M.; Kerr, D.A.; Thompson, P.L.; Stojceski, B.; Prince, R.L. The effect of yoghurt and its probiotics on blood pressure and serum lipid profile; a randomised controlled trial. Nutr. Metab. Cardiovasc. Dis. 2015, 25, 46–51. [Google Scholar] [CrossRef] [Green Version]
  91. Farhangi, M.A.; Javid, A.Z.; Dehghan, P. The effect of enriched chicory inulin on liver enzymes, calcium homeostasis and hematological parameters in patients with type 2 diabetes mellitus: A randomized placebo-controlled trial. Prim. Care Diabetes 2016, 10, 265–271. [Google Scholar] [CrossRef]
  92. Dehghan, P.; Farhangi, M.A.; Tavakoli, F.; Aliasgarzadeh, A.; Akbari, A.M. Impact of prebiotic supplementation on T-cell subsets and their related cytokines, anthropometric features and blood pressure in patients with type 2 diabetes mellitus: A randomized placebo-controlled Trial. Complement. Ther. Med. 2016, 24, 96–102. [Google Scholar] [CrossRef]
  93. Razmpoosh, E.; Javadi, A.; Ejtahed, H.S.; Mirmiran, P.; Javadi, M.; Yousefinejad, A. The effect of probiotic supplementation on glycemic control and lipid profile in patients with type 2 diabetes: A randomized placebo controlled trial. Diabetes Metab. Syndr. 2019, 13, 175–182. [Google Scholar] [CrossRef]
  94. Kobyliak, N.; Falalyeyeva, T.; Mykhalchyshyn, G.; Kyriienko, D.; Komissarenko, I. Effect of alive probiotic on insulin resistance in type 2 diabetes patients: Randomized clinical trial. Diabetes Metab. Syndr. 2018, 12, 617–624. [Google Scholar] [CrossRef] [PubMed]
  95. Mobini, R.; Tremaroli, V.; Ståhlman, M.; Karlsson, F.; Levin, M.; Ljungberg, M.; Sohlin, M.; Bertéus Forslund, H.; Perkins, R.; Bäckhed, F.; et al. Metabolic effects of Lactobacillus reuteri DSM 17938 in people with type 2 diabetes: A randomized controlled trial. Diabetes Obes. Metab. 2017, 19, 579–589. [Google Scholar] [CrossRef] [PubMed]
  96. Ostadrahimi, A.; Taghizadeh, A.; Mobasseri, M.; Farrin, N.; Payahoo, L.; Beyramalipoor Gheshlaghi, Z.; Vahedjabbari, M. Effect of probiotic fermented milk (kefir) on glycemic control and lipid profile in type 2 diabetic patients: A randomized double-blind placebo-controlled clinical trial. Iran. J. Public Health 2015, 44, 228–237. [Google Scholar] [PubMed]
  97. Shakeri, H.; Hadaegh, H.; Abedi, F.; Tajabadi-Ebrahimi, M.; Mazroii, N.; Ghandi, Y.; Asemi, Z. Consumption of synbiotic bread decreases triacylglycerol and VLDL levels while increasing HDL levels in serum from patients with type-2 diabetes. Lipids 2014, 49, 695–701. [Google Scholar] [CrossRef]
  98. Hosseinzadeh, P.; Javanbakht, M.H.; Mostafavi, S.A.; Djalali, M.; Derakhshanian, H.; Hajianfar, H.; Bahonar, A.; Djazayery, A. Brewer’s Yeast Improves Glycemic Indices in Type 2 Diabetes Mellitus. Int. J. Prev. Med. 2013, 4, 1131–1138. [Google Scholar]
  99. Asemi, Z.; Alizadeh, S.A.; Ahmad, K.; Goli, M.; Esmaillzadeh, A. Effects of beta-carotene fortified synbiotic food on metabolic control of patients with type 2 diabetes mellitus: A double-blind randomized cross-over controlled clinical trial. Clin. Nutr. 2016, 35, 819–825. [Google Scholar] [CrossRef]
  100. Dehghan, P.; Gargari, B.P.; Jafar-Abadi, M.A.; Aliasgharzadeh, A. Inulin controls inflammation and metabolic endotoxemia in women with type 2 diabetes mellitus: A randomized-controlled clinical trial. Int. J. Food Sci. Nutr. 2014, 65, 117–123. [Google Scholar] [CrossRef]
  101. Raygan, F.; Ostadmohammadi, V.; Asemi, Z. The effects of probiotic and selenium co-supplementation on mental health parameters and metabolic profiles in type 2 diabetic patients with coronary heart disease: A randomized, double-blind, placebo-controlled trial. Clin. Nutr. 2019, 38, 1594–1598. [Google Scholar] [CrossRef]
  102. Tenorio-Jiménez, C.; Martínez-Ramírez, M.J.; Gil, Á.; Gómez-Llorente, C. Effects of Probiotics on Metabolic Syndrome: A Systematic Review of Randomized Clinical Trials. Nutrients 2020, 12, 124. [Google Scholar] [CrossRef] [Green Version]
  103. Lee, E.; Jung, S.R.; Lee, S.Y.; Lee, N.K.; Paik, H.D.; Lim, S.I. Lactobacillus plantarum Strain Ln4 Attenuates Diet-Induced Obesity, Insulin Resistance, and Changes in Hepatic mRNA Levels Associated with Glucose and Lipid Metabolism. Nutrients 2018, 10, 643. [Google Scholar] [CrossRef] [Green Version]
  104. Zhang, Y.; Wang, L.; Zhang, J.; Li, Y.; He, Q.; Li, H.; Guo, X.; Guo, J.; Zhang, H. Probiotic Lactobacillus casei Zhang ameliorates high-fructose-induced impaired glucose tolerance in hyperinsulinemia rats. Eur. J. Nutr. 2014, 53, 221–232. [Google Scholar] [CrossRef]
  105. Toral, M.; Gómez-Guzmán, M.; Jiménez, R.; Romero, M.; Sánchez, M.; Utrilla, M.P.; Garrido-Mesa, N.; Rodríguez-Cabezas, M.E.; Olivares, M.; Gálvez, J.; et al. The probiotic Lactobacillus coryniformis CECT5711 reduces the vascular pro-oxidant and pro-inflammatory status in obese mice. Clin. Sci. 2014, 127, 33–45. [Google Scholar] [CrossRef]
  106. Hsieh, F.C.; Lan, C.C.; Huang, T.Y.; Chen, K.W.; Chai, C.Y.; Chen, W.T.; Fang, A.H.; Chen, Y.H.; Wu, C.S. Heat-killed and live Lactobacillus reuteri GMNL-263 exhibit similar effects on improving metabolic functions in high-fat diet-induced obese rats. Food Funct. 2016, 7, 2374–2388. [Google Scholar] [CrossRef] [Green Version]
  107. Lim, S.M.; Jeong, J.J.; Woo, K.H.; Han, M.J.; Kim, D.H. Lactobacillus sakei OK67 ameliorates high-fat diet-induced blood glucose intolerance and obesity in mice by inhibiting gut microbiota lipopolysaccharide production and inducing colon tight junction protein expression. Nutr. Res. 2016, 36, 337–348. [Google Scholar] [CrossRef] [PubMed]
  108. Li, X.; Wang, E.; Yin, B.; Fang, D.; Chen, P.; Wang, G.; Zhao, J.; Zhang, H.; Chen, W. Effects of Lactobacillus casei CCFM419 on insulin resistance and gut microbiota in type 2 diabetic mice. Benef. Microbes 2017, 8, 421–432. [Google Scholar] [CrossRef] [PubMed]
  109. Balakumar, M.; Prabhu, D.; Sathishkumar, C.; Prabu, P.; Rokana, N.; Kumar, R.; Raghavan, S.; Soundarajan, A.; Grover, S.; Batish, V.K.; et al. Improvement in glucose tolerance and insulin sensitivity by probiotic strains of Indian gut origin in high-fat diet-fed C57BL/6J mice. Eur. J. Nutr. 2018, 57, 279–295. [Google Scholar] [CrossRef] [PubMed]
  110. Singh, S.; Sharma, R.K.; Malhotra, S.; Pothuraju, R.; Shandilya, U.K. Lactobacillus rhamnosus NCDC17 ameliorates type-2 diabetes by improving gut function, oxidative stress and inflammation in high-fat-diet fed and streptozotocintreated rats. Benef. Microbes 2017, 8, 243–255. [Google Scholar] [CrossRef] [PubMed]
  111. Natividad, J.M.; Lamas, B.; Pham, H.P.; Michel, M.L.; Rainteau, D.; Bridonneau, C.; da Costa, G.; van Hylckama Vlieg, J.; Sovran, B.; Chamignon, C.; et al. Bilophila wadsworthia aggravates high fat diet induced metabolic dysfunctions in mice. Nat. Commun. 2018, 9, 2802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Niibo, M.; Shirouchi, B.; Umegatani, M.; Morita, Y.; Ogawa, A.; Sakai, F.; Kadooka, Y.; Sato, M. Probiotic Lactobacillus gasseri SBT2055 improves insulin secretion in a diabetic rat model. J. Dairy Sci. 2019, 102, 997–1006. [Google Scholar] [CrossRef] [Green Version]
  113. Wanchai, K.; Yasom, S.; Tunapong, W.; Chunchai, T.; Eaimworawuthikul, S.; Thiennimitr, P.; Chaiyasut, C.; Pongchaidecha, A.; Chatsudthipong, V.; Chattipakorn, S.; et al. Probiotic Lactobacillus paracasei HII01 protects rats against obese-insulin resistance-induced kidney injury and impaired renal organic anion transporter 3 function. Clin. Sci. 2018, 132, 1545–1563. [Google Scholar] [CrossRef] [PubMed]
  114. Kikuchi, K.; Ben Othman, M.; Sakamoto, K. Sterilized bifidobacteria suppressed fat accumulation and blood glucose level. Biochem. Biophys. Res. Commun. 2018, 501, 1041–1047. [Google Scholar] [CrossRef] [PubMed]
  115. Aoki, R.; Kamikado, K.; Suda, W.; Takii, H.; Mikami, Y.; Suganuma, N.; Hattori, M.; Koga, Y. A proliferative probiotic Bifidobacterium strain in the gut ameliorates progression of metabolic disorders via microbiota modulation and acetate elevation. Sci. Rep. 2017, 7, 43522. [Google Scholar] [CrossRef] [PubMed]
  116. Kondo, S.; Xiao, J.Z.; Satoh, T.; Odamaki, T.; Takahashi, S.; Sugahara, H.; Yaeshima, T.; Iwatsuki, K.; Kamei, A.; Abe, K. Antiobesity effects of Bifidobacterium breve strain B-3 supplementation in a mouse model with high-fat diet-induced obesity. Biosci. Biotechnol. Biochem. 2010, 74, 1656–1661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Amar, J.; Chabo, C.; Waget, A.; Klopp, P.; Vachoux, C.; Bermúdez-Humarán, L.G.; Smirnova, N.; Bergé, M.; Sulpice, T.; Lahtinen, S.; et al. Intestinal mucosal adherence and translocation of commensal bacteria at the early onset of type 2 diabetes: Molecular mechanisms and probiotic treatment. EMBO Mol. Med. 2011, 3, 559–572. [Google Scholar] [CrossRef] [PubMed]
  118. Shang, H.; Sun, J.; Chen, Y.Q. Clostridium Butyricum CGMCC0313.1 Modulates Lipid Profile, Insulin Resistance and Colon Homeostasis in Obese Mice. PLoS ONE 2016, 11, e0154373. [Google Scholar] [CrossRef] [PubMed]
  119. Thiennimitr, P.; Yasom, S.; Tunapong, W.; Chunchai, T.; Wanchai, K.; Pongchaidecha, A.; Lungkaphin, A.; Sirilun, S.; Chaiyasut, C.; Chattipakorn, N.; et al. Lactobacillus paracasei HII01, xylooligosaccharides, and synbiotics reduce gut disturbance in obese rats. Nutrition 2018, 54, 40–47. [Google Scholar] [CrossRef] [PubMed]
  120. Chunchai, T.; Thunapong, W.; Yasom, S.; Wanchai, K.; Eaimworawuthikul, S.; Metzler, G.; Lungkaphin, A.; Pongchaidecha, A.; Sirilun, S.; Chaiyasut, C.; et al. Decreased microglial activation through gut-brain axis by prebiotics, probiotics, or synbiotics effectively restored cognitive function in obese-insulin resistant rats. J. Neuroinflamm. 2018, 15, 11. [Google Scholar] [CrossRef] [PubMed]
  121. Morshedi, M.; Valenlia, K.B.; Hosseinifard, E.S.; Shahabi, P.; Abbasi, M.M.; Ghorbani, M.; Barzegari, A.; Sadigh-Eteghad, S.; Saghafi-Asl, M. Beneficial psychological effects of novel psychobiotics in diabetic rats: The interaction among the gut, blood and amygdala. J. Nutr. Biochem. 2018, 57, 145–152. [Google Scholar] [CrossRef]
  122. Yao, F.; Jia, R.; Huang, H.; Yu, Y.; Mei, L.; Bai, L.; Ding, Y.; Zheng, P. Effect of Lactobacillus paracasei N1115 and fructooligosaccharides in nonalcoholic fatty liver disease. Arch. Med. Sci. 2019, 15, 1336–1344. [Google Scholar] [CrossRef]
  123. Alard, J.; Lehrter, V.; Rhimi, M.; Mangin, I.; Peucelle, V.; Abraham, A.L.; Mariadassou, M.; Maguin, E.; Waligora-Dupriet, A.J.; Pot, B.; et al. Beneficial metabolic effects of selected probiotics on diet-induced obesity and insulin resistance in mice are associated with improvement of dysbiotic gut microbiota. Environ. Microbiol. 2016, 18, 1484–1497. [Google Scholar] [CrossRef]
  124. Bagarolli, R.A.; Tobar, N.; Oliveira, A.G.; Araújo, T.G.; Carvalho, B.M.; Rocha, G.Z.; Vecina, J.F.; Calisto, K.; Guadagnini, D.; Prada, P.O.; et al. Probiotics modulate gut microbiota and improve insulin sensitivity in DIO mice. J. Nutr. Biochem. 2017, 50, 16–25. [Google Scholar] [CrossRef] [Green Version]
  125. Xu, Y.; Wang, N.; Tan, H.Y.; Li, S.; Zhang, C.; Feng, Y. Function of Akkermansia muciniphila in Obesity: Interactions With Lipid Metabolism, Immune Response and Gut Systems. Front. Microbiol. 2020, 11. [Google Scholar] [CrossRef] [Green Version]
  126. Derrien, M.; Vaughan, E.E.; Plugge, C.M.; de Vos, W.M. Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int. J. Syst. Evol. Microbiol. 2004, 54, 1469–1476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Zhai, Q.; Feng, S.; Arjan, N.; Chen, W. A next generation probiotic, Akkermansia muciniphila. Crit. Rev. Food Sci. Nutr. 2019, 59, 3227–3236. [Google Scholar] [CrossRef]
  128. Huang, K.; Wang, M.; Kulinich, A.; Yao, H.; Ma, H.; Reyes Martínez, J.E.; Duan, X.; Chen, H.; Cai, Z.P.; Flitsch, S.; et al. Biochemical characterisation of the neuraminidase pool of the human gut symbiont Akkermansia muciniphila. Carbohydr. Res. 2015, 415, 60–65. [Google Scholar] [CrossRef]
  129. Ottman, N.; Huuskonen, L.; Reunanen, J.; Boeren, S.; Klievink, J.; Smidt, H.; Belzer, C.; de Vos, W.M. Characterization of Outer Membrane Proteome of Akkermansia muciniphila Reveals Sets of Novel Proteins Exposed to the Human Intestine. Front. Microbiol. 2016, 7, 1157. [Google Scholar] [CrossRef]
  130. Collado, M.C.; Derrien, M.; Isolauri, E.; de Vos, W.M.; Salminen, S. Intestinal integrity and Akkermansia muciniphila, a mucin-degrading member of the intestinal microbiota present in infants, adults, and the elderly. Appl. Environ. Microbiol. 2007, 73, 7767–7770. [Google Scholar] [CrossRef] [Green Version]
  131. Wang, L.; Christophersen, C.T.; Sorich, M.J.; Gerber, J.P.; Angley, M.T.; Conlon, M.A. Low relative abundances of the mucolytic bacterium Akkermansia muciniphila and Bifidobacterium spp. in feces of children with autism. Appl. Environ. Microbiol. 2011, 77, 6718–6721. [Google Scholar] [CrossRef] [Green Version]
  132. Swidsinski, A.; Dörffel, Y.; Loening-Baucke, V.; Theissig, F.; Rückert, J.C.; Ismail, M.; Rau, W.A.; Gaschler, D.; Weizenegger, M.; Kühn, S.; et al. Acute appendicitis is characterised by local invasion with Fusobacterium nucleatum/necrophorum. Gut 2011, 60, 34–40. [Google Scholar] [CrossRef]
  133. Dingemanse, C.; Belzer, C.; van Hijum, S.A.; Günthel, M.; Salvatori, D.; den Dunnen, J.T.; Kuijper, E.J.; Devilee, P.; de Vos, W.M.; van Ommen, G.B.; et al. Akkermansia muciniphila and Helicobacter typhlonius modulate intestinal tumor development in mice. Carcinogenesis 2015, 36, 1388–1396. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Hansen, C.H.; Krych, L.; Nielsen, D.S.; Vogensen, F.K.; Hansen, L.H.; Sørensen, S.J.; Buschard, K.; Hansen, A.K. Early life treatment with vancomycin propagates Akkermansia muciniphila and reduces diabetes incidence in the NOD mouse. Diabetologia 2012, 55, 2285–2294. [Google Scholar] [CrossRef] [Green Version]
  135. Pascale, A.; Marchesi, N.; Govoni, S.; Coppola, A.; Gazzaruso, C. The role of gut microbiota in obesity, diabetes mellitus, and effect of metformin: New insights into old diseases. Curr. Opin. Pharmacol. 2019, 49, 1–5. [Google Scholar] [CrossRef]
  136. Dao, M.C.; Everard, A.; Aron-Wisnewsky, J.; Sokolovska, N.; Prifti, E.; Verger, E.O.; Kayser, B.D.; Levenez, F.; Chilloux, J.; Hoyles, L.; et al. Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: Relationship with gut microbiome richness and ecology. Gut 2016, 65, 426–436. [Google Scholar] [CrossRef] [Green Version]
  137. Remely, M.; Hippe, B.; Geretschlaeger, I.; Stegmayer, S.; Hoefinger, I.; Haslberger, A. Increased gut microbiota diversity and abundance of Faecalibacterium prausnitzii and Akkermansia after fasting: A pilot study. Wien. Klin. Wochenschr. 2015, 127, 394–398. [Google Scholar] [CrossRef] [Green Version]
  138. Everard, A.; Matamoros, S.; Geurts, L.; Delzenne, N.M.; Cani, P.D. Saccharomyces boulardii administration changes gut microbiota and reduces hepatic steatosis, low-grade inflammation, and fat mass in obese and type 2 diabetic db/db mice. mBio 2014, 5, e01011-14. [Google Scholar] [CrossRef] [Green Version]
  139. Schneeberger, M.; Everard, A.; Gómez-Valadés, A.G.; Matamoros, S.; Ramírez, S.; Delzenne, N.M.; Gomis, R.; Claret, M.; Cani, P.D. Akkermansia muciniphila inversely correlates with the onset of inflammation, altered adipose tissue metabolism and metabolic disorders during obesity in mice. Sci. Rep. 2015, 5, 16643. [Google Scholar] [CrossRef] [Green Version]
  140. Brahe, L.K.; Le Chatelier, E.; Prifti, E.; Pons, N.; Kennedy, S.; Hansen, T.; Pedersen, O.; Astrup, A.; Ehrlich, S.D.; Larsen, L.H. Specific gut microbiota features and metabolic markers in postmenopausal women with obesity. Nutr. Diabetes 2015, 5, e159. [Google Scholar] [CrossRef]
  141. Everard, A.; Belzer, C.; Geurts, L.; Ouwerkerk, J.P.; Druart, C.; Bindels, L.B.; Guiot, Y.; Derrien, M.; Muccioli, G.G.; Delzenne, N.M.; et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc. Natl. Acad. Sci. USA 2013, 110, 9066–9071. [Google Scholar] [CrossRef] [Green Version]
  142. Everard, A.; Lazarevic, V.; Derrien, M.; Girard, M.; Muccioli, G.G.; Neyrinck, A.M.; Possemiers, S.; Van Holle, A.; François, P.; de Vos, W.M.; et al. Responses of gut microbiota and glucose and lipid metabolism to prebiotics in genetic obese and diet-induced leptin-resistant mice. Diabetes 2011, 60, 2775–2786. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Arora, A.; Behl, T.; Sehgal, A.; Singh, S.; Sharma, N.; Bhatia, T.; Sobarzo-Sanchez, E.; Bungau, S. Unravelling the Involvement of Gut Microbiota in Type 2 Diabetes Mellitus. Life Sci. 2021, 273, 119311. [Google Scholar] [CrossRef]
  144. Santacruz, A.; Collado, M.C.; García-Valdés, L.; Segura, M.T.; Martín-Lagos, J.A.; Anjos, T.; Martí-Romero, M.; Lopez, R.M.; Florido, J.; Campoy, C.; et al. Gut microbiota composition is associated with body weight, weight gain and biochemical parameters in pregnant women. Br. J. Nutr. 2010, 104, 83–92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Karlsson, C.L.; Onnerfält, J.; Xu, J.; Molin, G.; Ahrné, S.; Thorngren-Jerneck, K. The microbiota of the gut in preschool children with normal and excessive body weight. Obesity 2012, 20, 2257–2261. [Google Scholar] [CrossRef] [PubMed]
  146. Reunanen, J.; Kainulainen, V.; Huuskonen, L.; Ottman, N.; Belzer, C.; Huhtinen, H.; de Vos, W.M.; Satokari, R. Akkermansia muciniphila Adheres to Enterocytes and Strengthens the Integrity of the Epithelial Cell Layer. Appl. Environ. Microbiol. 2015, 81, 3655–3662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Rosario, D.; Benfeitas, R.; Bidkhori, G.; Zhang, C.; Uhlen, M.; Shoaie, S.; Mardinoglu, A. Understanding the Representative Gut Microbiota Dysbiosis in Metformin-Treated Type 2 Diabetes Patients Using Genome-Scale Metabolic Modeling. Front. Physiol. 2018, 9, 775. [Google Scholar] [CrossRef] [PubMed]
  148. Al Khodor, S.; Reichert, B.; Shatat, I.F. The Microbiome and Blood Pressure: Can Microbes Regulate Our Blood Pressure? Front. Pediatr. 2017, 5, 138. [Google Scholar] [CrossRef]
  149. Cani, P.D. Human gut microbiome: Hopes, threats and promises. Gut 2018, 67, 1716–1725. [Google Scholar] [CrossRef]
  150. Ottman, N.; Reunanen, J.; Meijerink, M.; Pietilä, T.E.; Kainulainen, V.; Klievink, J.; Huuskonen, L.; Aalvink, S.; Skurnik, M.; Boeren, S.; et al. Pili-like proteins of Akkermansia muciniphila modulate host immune responses and gut barrier function. PLoS ONE 2017, 12, e0173004. [Google Scholar] [CrossRef]
  151. Medina-Vera, I.; Sanchez-Tapia, M.; Noriega-López, L.; Granados-Portillo, O.; Guevara-Cruz, M.; Flores-López, A.; Avila-Nava, A.; Fernández, M.L.; Tovar, A.R.; Torres, N. A dietary intervention with functional foods reduces metabolic endotoxaemia and attenuates biochemical abnormalities by modifying faecal microbiota in people with type 2 diabetes. Diabetes Metab. 2019, 45, 122–131. [Google Scholar] [CrossRef]
  152. Roshanravan, N.; Mahdavi, R.; Alizadeh, E.; Ghavami, A.; Rahbar Saadat, Y.; Mesri Alamdari, N.; Alipour, S.; Dastouri, M.R.; Ostadrahimi, A. The effects of sodium butyrate and inulin supplementation on angiotensin signaling pathway via promotion of Akkermansia muciniphila abundance in type 2 diabetes; A randomized, double-blind, placebo-controlled trial. J. Cardiovasc. Thorac. Res. 2017, 9, 183–190. [Google Scholar] [CrossRef]
  153. Walker, J.M.; Eckardt, P.; Aleman, J.O.; da Rosa, J.C.; Liang, Y.; Iizumi, T.; Etheve, S.; Blaser, M.J.; Breslow, J.L.; Holt, P.R. The effects of trans-resveratrol on insulin resistance, inflammation, and microbiota in men with the metabolic syndrome: A pilot randomized, placebo-controlled clinical trial. J. Clin. Transl. Res. 2019, 4, 122–135. [Google Scholar] [PubMed]
  154. Allin, K.H.; Tremaroli, V.; Caesar, R.; Jensen, B.A.H.; Damgaard, M.T.F.; Bahl, M.I.; Licht, T.R.; Hansen, T.H.; Nielsen, T.; Dantoft, T.M.; et al. Aberrant intestinal microbiota in individuals with prediabetes. Diabetologia 2018, 61, 810–820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Khan, T.J.; Ahmed, Y.M.; Zamzami, M.A.; Siddiqui, A.M.; Khan, I.; Baothman, O.A.S.; Mehanna, M.G.; Kuerban, A.; Kaleemuddin, M.; Yasir, M. Atorvastatin Treatment Modulates the Gut Microbiota of the Hypercholesterolemic Patients. Omics 2018, 22, 154–163. [Google Scholar] [CrossRef] [PubMed]
  156. Liu, F.; Ling, Z.; Xiao, Y.; Lv, L.; Yang, Q.; Wang, B.; Lu, H.; Zheng, L.; Jiang, P.; Wang, W.; et al. Dysbiosis of urinary microbiota is positively correlated with type 2 diabetes mellitus. Oncotarget 2017, 8, 3798–3810. [Google Scholar] [CrossRef] [Green Version]
  157. Collado, M.C.; Isolauri, E.; Laitinen, K.; Salminen, S. Effect of mother’s weight on infant’s microbiota acquisition, composition, and activity during early infancy: A prospective follow-up study initiated in early pregnancy. Am. J. Clin. Nutr. 2010, 92, 1023–1030. [Google Scholar] [CrossRef]
  158. Collado, M.C.; Laitinen, K.; Salminen, S.; Isolauri, E. Maternal weight and excessive weight gain during pregnancy modify the immunomodulatory potential of breast milk. Pediatr. Res. 2012, 72, 77–85. [Google Scholar] [CrossRef] [Green Version]
  159. De la Cuesta-Zuluaga, J.; Mueller, N.T.; Corrales-Agudelo, V.; Velásquez-Mejía, E.P.; Carmona, J.A.; Abad, J.M.; Escobar, J.S. Metformin Is Associated With Higher Relative Abundance of Mucin-Degrading Akkermansia muciniphila and Several Short-Chain Fatty Acid-Producing Microbiota in the Gut. Diabetes Care 2017, 40, 54–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Palleja, A.; Kashani, A.; Allin, K.H.; Nielsen, T.; Zhang, C.; Li, Y.; Brach, T.; Liang, S.; Feng, Q.; Jørgensen, N.B.; et al. Roux-en-Y gastric bypass surgery of morbidly obese patients induces swift and persistent changes of the individual gut microbiota. Genome Med. 2016, 8, 67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Cortez, R.V.; Petry, T.; Caravatto, P.; Pessôa, R.; Sanabani, S.S.; Martinez, M.B.; Sarian, T.; Salles, J.E.; Cohen, R.; Taddei, C.R. Shifts in intestinal microbiota after duodenal exclusion favor glycemic control and weight loss: A randomized controlled trial. Surg. Obes. Relat. Dis. 2018, 14, 1748–1754. [Google Scholar] [CrossRef]
  162. Grander, C.; Adolph, T.E.; Wieser, V.; Lowe, P.; Wrzosek, L.; Gyongyosi, B.; Ward, D.V.; Grabherr, F.; Gerner, R.R.; Pfister, A.; et al. Recovery of ethanol-induced Akkermansia muciniphila depletion ameliorates alcoholic liver disease. Gut 2018, 67, 891–901. [Google Scholar] [CrossRef]
  163. Zhang, H.; DiBaise, J.K.; Zuccolo, A.; Kudrna, D.; Braidotti, M.; Yu, Y.; Parameswaran, P.; Crowell, M.D.; Wing, R.; Rittmann, B.E.; et al. Human gut microbiota in obesity and after gastric bypass. Proc. Natl. Acad. Sci. USA 2009, 106, 2365. [Google Scholar] [CrossRef] [Green Version]
  164. Weir, T.L.; Manter, D.K.; Sheflin, A.M.; Barnett, B.A.; Heuberger, A.L.; Ryan, E.P. Stool microbiome and metabolome differences between colorectal cancer patients and healthy adults. PLoS ONE 2013, 8, e70803. [Google Scholar] [CrossRef] [Green Version]
  165. 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]
  166. Escobar, J.S.; Klotz, B.; Valdes, B.E.; Agudelo, G.M. The gut microbiota of Colombians differs from that of Americans, Europeans and Asians. BMC Microbiol. 2014, 14, 311. [Google Scholar] [CrossRef] [Green Version]
  167. Puupponen-Pimiä, R.; Aura, A.M.; Oksman-Caldentey, K.M.; Myllärinen, P.; Saarela, M.; Mattila-Sandholm, T.; Poutanen, K. Development of functional ingredients for gut health. Trends Food Sci. Technol. 2002, 13, 3–11. [Google Scholar] [CrossRef]
  168. Farzaneh, V.; Gominho, J.; Pereira, H.; Carvalho, I.S. Screening of the antioxidant and enzyme inhibition potentials of Portuguese Pimpinella anisum L. seeds by GC-MS. Food Anal. Methods 2018, 11, 2645–2656. [Google Scholar] [CrossRef]
  169. Farzaneh, V.; Carvalho, I.S. A review of the health benefit potentials of herbal plant infusions and their mechanism of actions. Ind. Crop. Prod. 2015, 65, 247–258. [Google Scholar] [CrossRef]
  170. Manach, C.; Scalbert, A.; Morand, C.; Rémésy, C.; Jiménez, L. Polyphenols: Food sources and bioavailability. Am. J. Clin. Nutr. 2004, 79, 727–747. [Google Scholar] [CrossRef] [Green Version]
  171. Bowey, E.; Adlercreutz, H.; Rowland, I. Metabolism of isoflavones and lignans by the gut microflora: A study in germ-free and human flora associated rats. Food Chem. Toxicol. 2003, 41, 631–636. [Google Scholar] [CrossRef]
  172. Parkar, S.G.; Stevenson, D.E.; Skinner, M.A. The potential influence of fruit polyphenols on colonic microflora and human gut health. Int. J. Food Microbiol. 2008, 124, 295–298. [Google Scholar] [CrossRef]
  173. Selma, M.V.; Espín, J.C.; Tomás-Barberán, F.A. Interaction between phenolics and gut microbiota: Role in human health. J. Agric. Food Chem. 2009, 57, 6485–6501. [Google Scholar] [CrossRef]
  174. Rechner, A.R.; Smith, M.A.; Kuhnle, G.; Gibson, G.R.; Debnam, E.S.; Srai, S.K.; Moore, K.P.; Rice-Evans, C.A. Colonic metabolism of dietary polyphenols: Influence of structure on microbial fermentation products. Free Radic. Biol. Med. 2004, 36, 212–225. [Google Scholar] [CrossRef]
  175. Russell, W.; Duthie, G. Plant secondary metabolites and gut health: The case for phenolic acids. Proc. Nutr. Soc. 2011, 70, 389–396. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Kim, D.-H.; Jung, E.-A.; Sohng, I.-S.; Han, J.-A.; Kim, T.-H.; Han, M.J. Intestinal bacterial metabolism of flavonoids and its relation to some biological activities. Arch. Pharmacal. Res. 1998, 21, 17–23. [Google Scholar] [CrossRef] [PubMed]
  177. Monagas, M.; Khan, N.; Andrés-Lacueva, C.; Urpí-Sardá, M.; Vázquez-Agell, M.; Lamuela-Raventós, R.M.; Estruch, R. Dihydroxylated phenolic acids derived from microbial metabolism reduce lipopolysaccharide-stimulated cytokine secretion by human peripheral blood mononuclear cells. Br. J. Nutr. 2009, 102, 201–206. [Google Scholar] [CrossRef] [Green Version]
  178. Pashikanti, S.; de Alba, D.R.; Boissonneault, G.A.; Cervantes-Laurean, D. Rutin metabolites: Novel inhibitors of nonoxidative advanced glycation end products. Free Radic. Biol. Med. 2010, 48, 656–663. [Google Scholar] [CrossRef] [PubMed]
  179. Tuohy, K.M.; Conterno, L.; Gasperotti, M.; Viola, R. Up-regulating the human intestinal microbiome using whole plant foods, polyphenols, and/or fiber. J. Agric. Food Chem. 2012, 60, 8776–8782. [Google Scholar] [CrossRef]
  180. Duda-Chodak, A. The inhibitory effect of polyphenols on human gut microbiota. J. Physiol. Pharmacol. 2012, 63, 497–503. [Google Scholar]
  181. Cardona, F.; Andrés-Lacueva, C.; Tulipani, S.; Tinahones, F.J.; Queipo-Ortuño, M.I. Benefits of polyphenols on gut microbiota and implications in human health. J. Nutr. Biochem. 2013, 24, 1415–1422. [Google Scholar] [CrossRef] [Green Version]
  182. Chen, Y.; Li, Q.; Zhao, T.; Zhang, Z.; Mao, G.; Feng, W.; Wu, X.; Yang, L. Biotransformation and metabolism of three mulberry anthocyanin monomers by rat gut microflora. Food Chem. 2017, 237, 887–894. [Google Scholar] [CrossRef]
  183. Tao, J.-H.; Duan, J.-A.; Jiang, S.; Qian, Y.-Y.; Qian, D.-W. Biotransformation and metabolic profile of buddleoside with human intestinal microflora by ultrahigh-performance liquid chromatography coupled to hybrid linear ion trap/orbitrap mass spectrometer. J. Chromatogr. B 2016, 1025, 7–15. [Google Scholar] [CrossRef] [PubMed]
  184. Mayta-Apaza, A.C.; Pottgen, E.; De Bodt, J.; Papp, N.; Marasini, D.; Howard, L.; Abranko, L.; Van de Wiele, T.; Lee, S.O.; Carbonero, F. Impact of tart cherries polyphenols on the human gut microbiota and phenolic metabolites in vitro and in vivo. J. Nutr. Biochem. 2018, 59, 160–172. [Google Scholar] [CrossRef]
  185. Gowd, V.; Bao, T.; Chen, W. Antioxidant potential and phenolic profile of blackberry anthocyanin extract followed by human gut microbiota fermentation. Food Res. Int. 2019, 120, 523–533. [Google Scholar] [CrossRef] [PubMed]
  186. Theilmann, M.C.; Goh, Y.J.; Nielsen, K.F.; Klaenhammer, T.R.; Barrangou, R.; Abou Hachem, M. Lactobacillus acidophilus Metabolizes Dietary Plant Glucosides and Externalizes Their Bioactive Phytochemicals. mBio 2017, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Chen, W.; Su, H.; Xu, Y.; Bao, T.; Zheng, X. Protective effect of wild raspberry (Rubus hirsutus Thunb.) extract against acrylamide-induced oxidative damage is potentiated after simulated gastrointestinal digestion. Food Chem. 2016, 196, 943–952. [Google Scholar] [CrossRef] [PubMed]
  188. Ozdal, T.; Sela, D.A.; Xiao, J.; Boyacioglu, D.; Chen, F.; Capanoglu, E. The Reciprocal Interactions between Polyphenols and Gut Microbiota and Effects on Bioaccessibility. Nutrients 2016, 8, 78. [Google Scholar] [CrossRef]
  189. Sun, H.; Chen, Y.; Cheng, M.; Zhang, X.; Zheng, X.; Zhang, Z. The modulatory effect of polyphenols from green tea, oolong tea and black tea on human intestinal microbiota in vitro. J. Food Sci. Technol. 2018, 55, 399–407. [Google Scholar] [CrossRef] [PubMed]
  190. Kemperman, R.A.; Gross, G.; Mondot, S.; Possemiers, S.U.; Marzorati, M.; Van de Wiele, T.L.A.; Doré, J.; Vaughan, E.E. Impact of polyphenols from black tea and red wine/grape juice on a gut model microbiome. Food Res. Int. 2013, 53, 659–669. [Google Scholar] [CrossRef]
  191. Sánchez-Patán, F.; Cueva, C.; Monagas, M.; Walton, G.E.; Gibson, G.R.; Quintanilla-López, J.E.; Lebrón-Aguilar, R.; Martín-Álvarez, P.J.; Moreno-Arribas, M.V.; Bartolomé, B. In Vitro Fermentation of a Red Wine Extract by Human Gut Microbiota: Changes in Microbial Groups and Formation of Phenolic Metabolites. J. Agric. Food Chem. 2012, 60, 2136–2147. [Google Scholar] [CrossRef]
  192. Queipo-Ortuño, M.I.; Boto-Ordóñez, M.; Murri, M.; Gomez-Zumaquero, J.M.; Clemente-Postigo, M.; Estruch, R.; Cardona Diaz, F.; Andrés-Lacueva, C.; Tinahones, F.J. Influence of red wine polyphenols and ethanol on the gut microbiota ecology and biochemical biomarkers. Am. J. Clin. Nutr. 2012, 95, 1323–1334. [Google Scholar] [CrossRef] [PubMed]
  193. Cueva, C.; Sánchez-Patán, F.; Monagas, M.; Walton, G.E.; Gibson, G.R.; Martín-Álvarez, P.J.; Bartolomé, B.; Moreno-Arribas, M.V. In vitro fermentation of grape seed flavan-3-ol fractions by human faecal microbiota: Changes in microbial groups and phenolic metabolites. FEMS Microbiol. Ecol. 2013, 83, 792–805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Kim, Y.; Keogh, J.B.; Clifton, P.M. Polyphenols and Glycemic Control. Nutrients 2016, 8, 17. [Google Scholar] [CrossRef] [PubMed]
  195. Pallag, A.; Bungau, S.; Tit, D.M.; Jurca, T.; Sirbu, V.; Honiges, A.; Horhogea, C. Comparative Study of Polyphenols, Flavonoids and Chlorophylls in Equisetum arvense L. Populations. Rev. Chim. 2016, 67, 530–533. [Google Scholar]
  196. Copolovici, D.; Bungau, S.; Boscencu, R.; Tit, D.M.; Copolovici, L. The Fatty Acids Composition and Antioxidant Activity of Walnut Cold Press Oil. Rev. Chim. 2017, 68, 507–509. [Google Scholar] [CrossRef]
  197. Rastmanesh, R. High polyphenol, low probiotic diet for weight loss because of intestinal microbiota interaction. Chem. Biol. Interact. 2011, 189, 1–8. [Google Scholar] [CrossRef]
  198. Anhê, F.F.; Roy, D.; Pilon, G.; Dudonné, S.; Matamoros, S.; Varin, T.V.; Garofalo, C.; Moine, Q.; Desjardins, Y.; Levy, E.; et al. A polyphenol-rich cranberry extract protects from diet-induced obesity, insulin resistance and intestinal inflammation in association with increased Akkermansia spp. population in the gut microbiota of mice. Gut 2015, 64, 872–883. [Google Scholar] [CrossRef] [Green Version]
  199. Oprea, O.B.; Apostol, L.; Bungau, S.; Cioca, G.; Samuel, A.D.; Badea, M.; Gaceu, L. Researches on the Chemical Composition and the Rheological Properties of Wheat and Grape Epicarp Flour Mixes. Rev. Chim. 2018, 69, 70–75. [Google Scholar] [CrossRef]
  200. Roopchand, D.E.; Carmody, R.N.; Kuhn, P.; Moskal, K.; Rojas-Silva, P.; Turnbaugh, P.J.; Raskin, I. Dietary Polyphenols Promote Growth of the Gut Bacterium Akkermansia muciniphila and Attenuate High-Fat Diet-Induced Metabolic Syndrome. Diabetes 2015, 64, 2847–2858. [Google Scholar] [CrossRef] [Green Version]
  201. Kameyama, K.; Itoh, K. Intestinal colonization by a Lachnospiraceae bacterium contributes to the development of diabetes in obese mice. Microbes Environ. 2014, 29, 427–430. [Google Scholar] [CrossRef] [Green Version]
  202. Bungau, S.; Behl, T.; Tit, D.M.; Banica, F.; Bratu, O.G.; Diaconu, C.C.; Nistor-Cseppento, C.D.; Bustea, C.; Aron, R.A.C.; Vesa, C.M. Interactions between leptin and insulin resistance in patients with prediabetes, with and without NAFLD. Exp. Ther. Med. 2020, 20. [Google Scholar] [CrossRef] [PubMed]
  203. Mudaliar, S.; Henry, R.R.; Sanyal, A.J.; Morrow, L.; Marschall, H.U.; Kipnes, M.; Adorini, L.; Sciacca, C.I.; Clopton, P.; Castelloe, E.; et al. Efficacy and safety of the farnesoid X receptor agonist obeticholic acid in patients with type 2 diabetes and nonalcoholic fatty liver disease. Gastroenterology 2013, 145, 574–582.e571. [Google Scholar] [CrossRef]
  204. Vesa, C.M.; Behl, T.; Nemeth, S.; Bratu, O.G.; Diaconu, C.C.; Moleriu, R.D.; Negrut, N.; Zaha, D.C.; Bustea, C.; Radu, F.I.; et al. Prediction of NAFLD occurrence in prediabetes patients. Exp. Ther. Med. 2020, 20. [Google Scholar] [CrossRef]
  205. Kuhn, P.; Kalariya, H.M.; Poulev, A.; Ribnicky, D.M.; Jaja-Chimedza, A.; Roopchand, D.E.; Raskin, I. Grape polyphenols reduce gut-localized reactive oxygen species associated with the development of metabolic syndrome in mice. PLoS ONE 2018, 13, e0198716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Shin, N.R.; Lee, J.C.; Lee, H.Y.; Kim, M.S.; Whon, T.W.; Lee, M.S.; Bae, J.W. An increase in the Akkermansia spp. population induced by metformin treatment improves glucose homeostasis in diet-induced obese mice. Gut 2014, 63, 727–735. [Google Scholar] [CrossRef] [Green Version]
  207. Luck, H.; Tsai, S.; Chung, J.; Clemente-Casares, X.; Ghazarian, M.; Revelo, X.S.; Lei, H.; Luk, C.T.; Shi, S.Y.; Surendra, A.; et al. Regulation of obesity-related insulin resistance with gut anti-inflammatory agents. Cell Metab. 2015, 21, 527–542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Baldwin, J.; Collins, B.; Wolf, P.G.; Martinez, K.; Shen, W.; Chuang, C.C.; Zhong, W.; Cooney, P.; Cockrell, C.; Chang, E.; et al. Table grape consumption reduces adiposity and markers of hepatic lipogenesis and alters gut microbiota in butter fat-fed mice. J. Nutr. Biochem. 2016, 27, 123–135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Pierre, J.F.; Heneghan, A.F.; Feliciano, R.P.; Shanmuganayagam, D.; Roenneburg, D.A.; Krueger, C.G.; Reed, J.D.; Kudsk, K.A. Cranberry proanthocyanidins improve the gut mucous layer morphology and function in mice receiving elemental enteral nutrition. JPEN J. Parenter Enter. Nutr. 2013, 37, 401–409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Etxeberria, U.; Arias, N.; Boqué, N.; Macarulla, M.T.; Portillo, M.P.; Martínez, J.A.; Milagro, F.I. Reshaping faecal gut microbiota composition by the intake of trans-resveratrol and quercetin in high-fat sucrose diet-fed rats. J. Nutr. Biochem. 2015, 26, 651–660. [Google Scholar] [CrossRef]
  211. Li, Z.; Henning, S.M.; Lee, R.P.; Lu, Q.Y.; Summanen, P.H.; Thames, G.; Corbett, K.; Downes, J.; Tseng, C.H.; Finegold, S.M.; et al. Pomegranate extract induces ellagitannin metabolite formation and changes stool microbiota in healthy volunteers. Food Funct. 2015, 6, 2487–2495. [Google Scholar] [CrossRef] [PubMed]
  212. Shen, W.; Shen, M.; Zhao, X.; Zhu, H.; Yang, Y.; Lu, S.; Tan, Y.; Li, G.; Li, M.; Wang, J.; et al. Anti-obesity Effect of Capsaicin in Mice Fed with High-Fat Diet Is Associated with an Increase in Population of the Gut Bacterium Akkermansia muciniphila. Front. Microbiol. 2017, 8, 272. [Google Scholar] [CrossRef] [PubMed]
  213. Das, S.S.; Bharadwaj, P.; Bilal, M.; Barani, M.; Rahdar, A.; Taboada, P.; Bungau, S.; Kyzas, G.Z. Stimuli-Responsive Polymeric Nanocarriers for Drug Delivery, Imaging, and Theragnosis. Polymers 2020, 12, 1397. [Google Scholar] [CrossRef] [PubMed]
  214. Karimi, M.; Yazdi, F.T.; Mortazavi, S.A.; Shahabi-Ghahfarrokhi, I.; Chamani, J. Development of active antimicrobial poly (l-glutamic) acid-poly (l-lysine) packaging material to protect probiotic bacterium. Polym. Test. 2020, 83, 106338. [Google Scholar] [CrossRef]
  215. Atia, A.; Gomaa, A.; Fliss, I.; Beyssac, E.; Garrait, G.; Subirade, M. A prebiotic matrix for encapsulation of probiotics: Physicochemical and microbiological study. J. Microencapsul. 2016, 33, 89–101. [Google Scholar] [CrossRef] [PubMed]
  216. Sozer, N.; Kokini, J.L. Nanotechnology and its applications in the food sector. Trends Biotechnol. 2009, 27, 82–89. [Google Scholar] [CrossRef]
  217. Felisberto, M.H.F.; Galvão, M.T.E.L.; Picone, C.S.F.; Cunha, R.L.; Pollonio, M.A.R. Effect of prebiotic ingredients on the rheological properties and microstructure of reduced-sodium and low-fat meat emulsions. LWT Food Sci. Technol. 2015, 60, 148–155. [Google Scholar] [CrossRef] [Green Version]
  218. Kazmierczak, R.; Choe, E.; Sinclair, J.; Eisenstark, A. Direct attachment of nanoparticle cargo to Salmonella typhimurium membranes designed for combination bacteriotherapy against tumors. Methods Mol. Biol. 2015, 1225, 151–163. [Google Scholar] [CrossRef]
  219. Hu, Q.; Wu, M.; Fang, C.; Cheng, C.; Zhao, M.; Fang, W.; Chu, P.K.; Ping, Y.; Tang, G. Engineering nanoparticle-coated bacteria as oral DNA vaccines for cancer immunotherapy. Nano Lett. 2015, 15, 2732–2739. [Google Scholar] [CrossRef]
  220. Feher, J.; Pinter, E.; Helyes, Z.; Szolcsanyi, J. Nano-size Particles Of Probiotics For Preventing And Treating Neuroinflammation. Investig. Ophthalmol. Vis. Sci. 2012, 53, 331. [Google Scholar]
  221. Kim, W.S.; Han, G.G.; Hong, L.; Kang, S.K.; Shokouhimehr, M.; Choi, Y.J.; Cho, C.S. Novel production of natural bacteriocin via internalization of dextran nanoparticles into probiotics. Biomaterials 2019, 218, 119360. [Google Scholar] [CrossRef]
  222. Kim, W.S.; Lee, J.Y.; Singh, B.; Maharjan, S.; Hong, L.; Lee, S.M.; Cui, L.H.; Lee, K.J.; Kim, G.; Yun, C.H.; et al. A new way of producing pediocin in Pediococcus acidilactici through intracellular stimulation by internalized inulin nanoparticles. Sci. Rep. 2018, 8, 5878. [Google Scholar] [CrossRef]
  223. Hong, L.; Kim, W.S.; Lee, S.M.; Kang, S.K.; Choi, Y.J.; Cho, C.S. Pullulan Nanoparticles as Prebiotics Enhance the Antibacterial Properties of Lactobacillus plantarum Through the Induction of Mild Stress in Probiotics. Front. Microbiol. 2019, 10, 142. [Google Scholar] [CrossRef] [Green Version]
  224. Song, Q.; Zheng, C.; Jia, J.; Zhao, H.; Feng, Q.; Zhang, H.; Wang, L.; Zhang, Z.; Zhang, Y. A Probiotic Spore-Based Oral Autonomous Nanoparticles Generator for Cancer Therapy. Adv. Mater. 2019, 31, e1903793. [Google Scholar] [CrossRef] [PubMed]
  225. Aziz Mousavi, S.M.A.; Mirhosseini, S.A.; Rastegar Shariat Panahi, M.; Mahmoodzadeh Hosseini, H. Characterization of Biosynthesized Silver Nanoparticles Using Lactobacillus rhamnosus GG and its In Vitro Assessment Against Colorectal Cancer Cells. Probiotics Antimicrob. Proteins 2020, 12, 740–746. [Google Scholar] [CrossRef] [PubMed]
  226. Fung, W.Y.; Yuen, K.H.; Liong, M.T. Agrowaste-based nanofibers as a probiotic encapsulant: Fabrication and characterization. J. Agric. Food Chem. 2011, 59, 8140–8147. [Google Scholar] [CrossRef]
  227. Ebrahimnejad, P.; Khalili, S.; Khavari, M. Survival of Lactobacillus Acidophilus as Probiotic Bacteria using Chitosan Nanoparticles. Int. J. Eng. 2017, 30. [Google Scholar] [CrossRef]
  228. Krithika, B.; Preetha, R. Formulation of protein based inulin incorporated synbiotic nanoemulsion for enhanced stability of probiotic. Mater. Res. Express 2019, 6, 114003. [Google Scholar] [CrossRef]
  229. Salmerón, I. Fermented cereal beverages: From probiotic, prebiotic and synbiotic towards Nanoscience designed healthy drinks. Lett. Appl. Microbiol. 2017, 65, 114–124. [Google Scholar] [CrossRef] [Green Version]
  230. Guimarães, J.T.; Balthazar, C.F.; Scudino, H.; Pimentel, T.C.; Esmerino, E.A.; Ashokkumar, M.; Freitas, M.Q.; Cruz, A.G. High-intensity ultrasound: A novel technology for the development of probiotic and prebiotic dairy products. Ultrason. Sonochem. 2019, 57, 12–21. [Google Scholar] [CrossRef]
Figure 1. Flow chart of the selection process of the bibliographic sources included in this article.
Figure 1. Flow chart of the selection process of the bibliographic sources included in this article.
Microorganisms 09 00618 g001
Figure 2. A. muciniphila’s benefits in MetS.
Figure 2. A. muciniphila’s benefits in MetS.
Microorganisms 09 00618 g002
Table 1. Summarizing the published data related to the clinical trials considering the effects of pre-/probiotics on the associated risk factors to MetS development.
Table 1. Summarizing the published data related to the clinical trials considering the effects of pre-/probiotics on the associated risk factors to MetS development.
Type of the Controlled TrialForm of the Product; Species/Active Substance; Period of Administration, Dose of Pre/ProbioticEffects/ActionsRef.
Dyslipidemia
Randomized double-blind placebo, crossoverLyophilized probiotic culture capsules; Bifidobacterium animalis ssp. lactis MB 2409 (DSM 23733) + Bifidobacterium MB 109 (DSM 23731) + Bifidobacterium longum ssp. longum BL04 (DSM 23233) (109 CFU/g); 12 weeks, 1/day↓: LDL-C, TC
↑: HDL-C
[72]
Randomized double-blind placeboLyophilized symbiotic capsules; Lactobacillus acidophilus CHO-220 (109 CFU/g) + 0.2 g Inulin; 12 weeks, 4/day↓: LDL-C, TC[73]
Hypercholesterolemia
Single-arm, open-label pilot studyProbiotic culture capsules; Saccharomyces cerevisiae var. boulardii CNCM I-1079 (1.4 × 1010 CFU); 8 weeks, 2/day↓: RLP-P[74]
Hypertensive adults
Double-blind placeboFruit drink with probiotic bacteria; Lactobacillus plantarum DSM 15313; 1 × 109 CFU/day dose; 12 weeks, 1/dayNot effective: BP parameters[75]
Yogurt; Enterococcus faecium + Streptococcus thermophilus 4.7 × 1011; 8 weeks↓: SBP, DBP[76]
Sour milk; Lactobacillus helveticus + Saccharomyces cerevisiae; 7 × 1010; 8 weeks[77]
Capsules; Lactobacillus reuteri 5.8 × 109; 8 weeks[78]
Cheese; L. plantarum 7.5 × 1012; 3 weeks[79]
Hypertensive overweight women
Double blind, randomizedCheese with probiotic bacteria/Lactobacillus casei 01 (108 CFU/g); 4 weeks, 1/day↓: TC, LDL-C, TG, SBP, DBP
↑: HB, HDL-C, HE
[80]
Metabolic syndrome
Randomized double-blind placeboPre/probiotic culture in yogurt/milk; Bifidobacterium lactis Bb-12 (107 CFU/g) + 6 g inulin; 10 weeks, 2/day↓: BFM, BFP, HOMA-IR, TG, WC,
↑: 25(OH)D, HDL-C, QUICKI
[81]
Pro/synbiotics culture in packages; Lactobacillus acidophilus + Bifidobacterium bifidum + Bifidobacterium lactis + Bifidobacterium longum (1.5 × 109 each); 24 weeks, 6 g/day
Observation: synbiotics (containing the above-mentioned probiotics) + inulin (prebiotic)
↓: HDL-C (probiotic group)[82]
Probiotic culture in yogurt/milk; Bifidobacterium lactis Bb-12 (3.6 × 106 CFU/300 g) + Lactobacillus acidophilus La-5 (4.4 × 106 CFU/300 g); 8 weeks, 1/day↓: BG, INS, HOMA-IR
↑: QUICKI
[83]
Milk; Lactobacillus casei; 108 cells/mL, 65 mL bottles × 3/day, 12 weeks ↑: High-sensitive CRP (1.86 mg/L (probiotic group) vs. −1.60 mg/L (placebo group), p = 0.016); LBP levels (5827 ng/mL (probiotic group) vs. −1510 ng/mL (placebo group), p = 0.023)[84]
Cheese; Lactobacillus plantarum; 1.5 × 1011 CFU/g, 50 g/day, 12 weeks↓: BMI (−2 (probiotic group) vs. −1.6 kg/m2 (placebo group), p = 0.031[79]
Milk; Lactobacillus casei; 108 cells/mL, 65 mL bottles × 3/day, 12 weeks↓: sVCAM-1 level (−195 ng/mL (probiotic group) vs. 30 ng/mL (placebo group), p = 0.008
↑: high-sensitive CRP level (1.86 mg/L (probiotic group) vs. −1.60 mg/L (placebo group), p = 0.002
[85]
Milk; Lactobacillus plantarum; 107 CFU/g, 80 mL bottles × 1/day, 12 weeks↓: Glucose levels in FM group vs. NFM group (−10.5 (FM group) vs. −3 mg/dL (NFM group), p = 0.037[86]
Milk; Bifidobacterium lactis; 3.4 × 108 CFU/mL, 80 mL bottle × 1/day, 6 weeks↓: BMI variation (−1.3 (probiotic group) vs. −0.3 kg/m2 (control group) p = 0.017); TC (−15 (probiotic group) vs. 6 mg/dL (control group), p = 0.09); LDL-C (−17.5 (probiotic group) vs. −2 mg/dL (control group), p = 0.08)[87]
Randomized double-blind placebo, cluster cross-overProbiotic culture in fermented milk;
Bifidobacterium animalis ssp. lactis HN019; 45 days, 1/day, 2.72 × 1010 CFU/mL
↓: BMI, TC, TNF-α, IL-6, LDL-C[87]
Overweight/obesity
Randomized double-blind placeboPrebiotic packages; 0.29 g oligofructose/kg body weight + 0.14 g oligofructose/kg body weight; 120 days, 1/day ↓: BMI, BW, INS, HOMA-IR, LDL-C, WC [88]
Lyophilized probiotic culture capsules;
Lactobacillus gasseri BNR17; 12 weeks, 2/day, 400 mg/capsule of low dose (109 CFU) or high dose (1010 CFU);
↓: VAT (high dose), WC in both dose (high and low)[89]
Prebiotic packages; 2 g oligofructose; 12 weeks, 1/day↓: BG, BW, INS[90]
Randomized, controlled, parallel, double-blind, factorial trialLyophilized probiotic cultures yoghurt/capsules; Lactobacillus acidophilus La-5 (3 × 109 CFU) + Bifidobacterium animalis Bb-12 (3 × 109 CFU); 6 weeks, 1/dayNot effective: lipid profile, BP[90]
T2DM
Randomized double-blind placeboPrebiotic packages; 10 g chicory inulin + oligofructose; 8 weeks, 1/day↓: AST, ALP, DBP, FSG, HbA1c, SBP, SC [91]
Prebiotic packages; 10 g oligofructose + inulin; 8 weeks, 1/day↓: BMI, DBP, IFN-γ, IL-12, WC,
↑: IL-4
[92]
Symbiotic packages/Lactobacillus acidophilus
(2 × 109 CFU) + Lactobacillus casei (7 × 109 CFU)
+ Lactobacillus bulgaricus (2 × 108 CFU) + Lactobacillus rhamnosus (1.5 × 109 CFU) + Bifidobacterium breve (3 × 1010 CFU) + Bifidobacterium longum
(7 × 109 CFU) + Streptococcus thermophilus
(1.5 × 109 CFU) + 100 mg oligofructose; 6 weeks, 1/day
↓: FPG
↑: HDL-C
[93]
Symbiotic packages; Lactobacillus + Bifidobacterium (1010 CFU/g) + Lactococcus (6 × 1010 CFU/g) + Propionibacterium (3 × 1010 CFU/g) + Acetobacter (1 × 106 CFU/g); 8 weeks, 1/day↓: HOMA-IR, IL-1β, TNF-α[94]
Probiotic yogurt; L. acidophilus La-5, B. lactis Bb-12; 8 weeks, 300 g/day, 3.7 × 106 CFU/mg for both organisms↓: FBG, HbA1c [95]
Dietary supplements, Lactobacillus reuteri DSM 17938; 12 weeks, low dose (108 CFU)/high dose (1010 CFU), ↓: HbA1c, insulin sensitivity index [11]
Probiotic fermented milk; Lactobacillus casei, L. acidophilus, bifidobacterial; 8 weeks, 300 mL/day↓: FBG, HbA1c [96]
Probiotic bread; Lactobacillus sporogenes; 8 weeks, 120 g/day, 1 × 108 CFU/g)↓: FBG, insulin, HOMA-IR, HOMA–B, QUICKI [97]
Tablets; Saccharomyces cerevisiae; 12 weeks, 1800 mg/day = 6 tablets↓: FBG, HbA1c, HOMA- IR, QUICKI, [98]
Randomized double-blind placebo crossoverSynbiotic packages; Lactobacillus sporogenes
(107 CFU/g) + 0.05 g β-carotene + 0.1 g inulin; 6 weeks, 3/day
↓: INS, HOMA-B, HOMA-IR, TC/HDL-C ratio, TG, VLDL-C [99]
Randomized triple-blind placeboPrebiotic packages; 10 g Inulin; 8 weeks, 1/day↓: FSG, HbA1c, HOMA-IR, hs-CRP, INS, TNF-α, LPS [100]
T2DM + CHD
Randomized double-blind placeboProbiotics culture and Se packages;
200 μg/day Se + 8 × 109 CFU/g probiotic (Lactobacillus acidophilus + Lactobacillus reuteri + Lactobacillus fermentum + Bifidobacterium bifidum (2 × 109 CFU/g each); 12 weeks, 1/day
↓: FPG, HOMA-IR (probiotic + Se), hs-CRP, INS, TC, TG, VLDL-C,
↑: GSH, NO, TAC, (co-supplementation)
[101]
Legend: ↑—increase;↓—reduction/decrease; 25(OH)D—25-hydroxyvitamin D; ALP—alkaline phosphatase; AST—aspartate aminotransferase; AV—abdominal visceral; BFM—body fat mass; BFP—body fat percentage; BG—blood glucose; BMI—body mass index measure; BP—blood pressure; BW—body weight; CFU—colony forming unit; CHD—coronary heart disease; DBP—diastolic blood pressure; FPG—fasting plasma glucose; FSG—fasting serum/blood glucose; GPA—glutathione peroxidase activities; GSH—total glutathione; HB—hemoglobin; HbA1c—glycated hemoglobin; HDL-C—high-density lipoprotein cholesterol; HE—hematocrit; HOMA-B—homeostasis model assessment index-β-cell; HOMA-IR—homeostasis model assessment index-insulin resistance; hs-CRP—high-sensitive C-reactive protein; IFN-γ—interferons-γ; IL-1β—interleukin-1β; IL-2—interleukin-2; IL-4—Interleukin-4; INS—insulin; LDL-C—low-density lipoprotein cholesterol; LPS—lipopolysaccharides; NO—nitric oxide; QUICKI—quantitative insulin sensitivity check index; RLP-P—remnant lipoprotein particle; SBP—systolic blood pressure; SC—serum calcium; Se—selenium; SFA—subcutaneous fat areas; T2DM—type 2 diabetes mellitus; TAC—total antioxidant capacity; TAS—total antioxidant status; TC—total cholesterol; TG—triglycerides; TNF-α—tumor necrosis factor alpha; VAT—visceral adipose tissue; VLDL-C—very low-density lipoprotein cholesterol; WC—waist circumference.
Table 2. Clinical studies of obesity implying A. muciniphila and its associated biomarkers.
Table 2. Clinical studies of obesity implying A. muciniphila and its associated biomarkers.
Patients Included in the Study/Condition/Period of StudyObservationsResultsRef.
81/T2DM/3 monthsA reduced-energy dietConsumption of A. muciniphila according to dietary portfolio improved levels and strengthened glycemic regulation, dyslipidemia, and inflammation.[151]
60/overweight + obese diabetes/45 days600 mg butyrate + 10 g inulin/day powder, or placeboInsulin + butyrate supplementation may increase A. muciniphila, and butyrate lowers the expression of TNF-alpha mRNA, hs-CRP, MDA, and DBP.[152]
28 men/obese + metabolic syndrome/35 days1 g resveratrol orally, 2/day or placeboResveratrol increases homeostasis of glucose and A. muciniphila abundance.[153]
134 prediabetes
134 healthy controls
ObservationThere was a strong decline in the concentration of the mucin-degrading bacterium A. muciniphila in prediabetes.[154]
49/overweight + obese/12 weeksCalorie restriction for 6 weeksThe large amounts of A. muciniphila increased the distribution of fasting plasma glucose, plasma triglycerides, and body fat.[136]
43/hypercholesterolemic 19 healthy controls/2 years27 patients with Atorvastatin treatmentTreatment with atorvastatin improved the amount of A. muciniphila.[155]
70 female patients/T2DM
70 healthy females.
ObservationDecreased A. muciniphila was linked with fasting blood/urinary glucose.[156]
16 infants/obese mothers 256 infants/normal mothers as controlObservationPrevalence of A. muciniphila was lower in control infants with normal mothers.[144,157,158]
28/diabetes
84 healthy controls
MetforminPatients with diabetes who obtained metformin have a higher relative abundance of A. muciniphila vs. healthy controls.[159]
13/morbidly obese patients/12 monthsRoux-en-Y gastric bypass (RYGB)Within the first 3 months, RYGB modified the relative abundances of 31 species, including A. muciniphila. This increase in abundance can be continued for 9 months.[160]
53 women/obesityObservationThere were 140 metagenomic species associated with metabolic markers, including A. muciniphila.[140]
21/T2DM/12 monthsDuodenal-jejunal by-pass surgery medical careIn the surgery control group, the amount of gut A. muciniphila increased.[57,161]
32/overweight + obese insulin-resistant/3 monthsOral supplementation of 1010 A. muciniphila bacteria, live or pasteurizedFor liver dysfunction and inflammation, A. muciniphila decreased body weight and decreased the levels of the related blood markers, although the overall composition of the gut microbiota remained unchanged.[59]
21/alcoholic steatohepatitis
16/healthy controls
ObservationThe concentration of fecal A. muciniphila vs. healthy controls (indirectly related to the incidence of hepatic disease) was diminished in ASH patients.[162]
13/overweight adults/7 weeksInterventional, fasting (1 week), followed by probiotic intake (6 weeks)
Samples: feces, T1 = before fasting, T2 = during fasting, T3 = 6 weeks after probiotic intervention
In comparison to fasting (T2), the concentration of A. muciniphila was higher before fasting (T1) than after probiotic action (T3)[137]
3c/normal weight
3/morbid obesity
3/after gastric bypass surgery
Interventional, gastric bypassReduced quantity of A. muciniphila in obese subjects (who obtained an elevated quantity of A. muciniphila after gastric bypass) vs. average weight subjects[163]
11/colorectal cancer
10/healthy
ObservationalIncreased A. muciniphila level in colorectal cancer patients vs. in healthy subjects[164]
71/T2DM
74/healthy controls
Increased A. muciniphila
abundance in T2DM patients’ feces, vs. in healthy controls patients
[165]
53/obese womenNo correlation of A. muciniphila abundance with dyslipidemia/insulin resistance[140]
Children (4–5-year-old)
20/normal weight
20/overweight
20/obesity
Reduced A. muciniphila abundance in overweight/obese vs. in normal weight children[145]
Pre-diabetes/newly diagnosed T2DM:
44/normal
64/pre-diabetes
13/T2DM
Reduced A. muciniphila abundance in patients with pre-diabetes/T2DM vs. subjects with regular glucose resistance[56]
23/autistic childrenReduced A. muciniphila abundance in autistic children’ feces[131]
Overweight/obese adults
10/normal weight
10/overweight
10/obesity
No correlation of A. muciniphila level had with the value of BMI[166]
Legend: BMI—body mass index; DPB—diastolic blood pressure; T2DM—type 2 diabetes mellitus.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Corb Aron, R.A.; Abid, A.; Vesa, C.M.; Nechifor, A.C.; Behl, T.; Ghitea, T.C.; Munteanu, M.A.; Fratila, O.; Andronie-Cioara, F.L.; Toma, M.M.; et al. Recognizing the Benefits of Pre-/Probiotics in Metabolic Syndrome and Type 2 Diabetes Mellitus Considering the Influence of Akkermansia muciniphila as a Key Gut Bacterium. Microorganisms 2021, 9, 618. https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms9030618

AMA Style

Corb Aron RA, Abid A, Vesa CM, Nechifor AC, Behl T, Ghitea TC, Munteanu MA, Fratila O, Andronie-Cioara FL, Toma MM, et al. Recognizing the Benefits of Pre-/Probiotics in Metabolic Syndrome and Type 2 Diabetes Mellitus Considering the Influence of Akkermansia muciniphila as a Key Gut Bacterium. Microorganisms. 2021; 9(3):618. https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms9030618

Chicago/Turabian Style

Corb Aron, Raluca Anca, Areha Abid, Cosmin Mihai Vesa, Aurelia Cristina Nechifor, Tapan Behl, Timea Claudia Ghitea, Mihai Alexandru Munteanu, Ovidiu Fratila, Felicia Liana Andronie-Cioara, Mirela Marioara Toma, and et al. 2021. "Recognizing the Benefits of Pre-/Probiotics in Metabolic Syndrome and Type 2 Diabetes Mellitus Considering the Influence of Akkermansia muciniphila as a Key Gut Bacterium" Microorganisms 9, no. 3: 618. https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms9030618

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