Next Article in Journal
Non-Markov-Type Analysis and Diffusion Map Analysis for Molecular Dynamics Trajectory of Chignolin at a High Temperature
Next Article in Special Issue
Aeromonas allosaccharophila Strain AE59-TE2 Is Highly Antagonistic towards Multidrug-Resistant Human Pathogens, What Does Its Genome Tell Us?
Previous Article in Journal
Modularity of the Human Musculoskeletal System: The Correlation between Functional Structures by Computer Tools Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Probiotic Mechanisms Affecting Glucose Homeostasis: A Scoping Review

Department of Microbiology, Biochemistry, Molecular Biology and Biotechnology, Faculty of Agriculture and Life Sciences, University of Maribor, Pivola 10, 2311 Hoče, Slovenia
*
Author to whom correspondence should be addressed.
Life 2022, 12(8), 1187; https://doi.org/10.3390/life12081187
Submission received: 19 June 2022 / Revised: 28 July 2022 / Accepted: 29 July 2022 / Published: 3 August 2022
(This article belongs to the Collection Feature Papers in Microbiology)

Abstract

:
The maintenance of a healthy status depends on the coexistence between the host organism and the microbiota. Early studies have already focused on the nutritional properties of probiotics, which may also contribute to the structural changes in the gut microbiota, thereby affecting host metabolism and homeostasis. Maintaining homeostasis in the body is therefore crucial and is reflected at all levels, including that of glucose, a simple sugar molecule that is an essential fuel for normal cellular function. Despite numerous clinical studies that have shown the effect of various probiotics on glucose and its homeostasis, knowledge about the exact function of their mechanism is still scarce. The aim of our review was to select in vivo and in vitro studies in English published in the last eleven years dealing with the effects of probiotics on glucose metabolism and its homeostasis. In this context, diverse probiotic effects at different organ levels were highlighted, summarizing their potential mechanisms to influence glucose metabolism and its homeostasis. Variations in results due to different methodological approaches were discussed, as well as limitations, especially in in vivo studies. Further studies on the interactions between probiotics, host microorganisms and their immunity are needed.

1. Introduction

Glucose is one of the simple sugars (monosaccharides) and as such it represents the main fuel for the normal functioning of the cell [1]. Maintaining of glucose homeostasis [2] is therefore essential to prevent the development of diseases associated with a disturbance of glucose homeostasis, such as insulin resistance [3], glucose intolerance [4] and type 2 diabetes [5]. The pathological condition common to all three is the occurrence of an elevated blood glucose level, i.e., hyperglycaemia [6]. The condition of persistent hyperglycaemia is very dangerous and leads to more serious complications associated with heart disease, eye, kidney and nerve damage [7], as well as with cancer [8]. Although the aetiology of hyperglycaemia has not been clearly understood, the main causes are attributed to genetic nature and/or to environmental factors [9,10]. The latter include an unhealthy lifestyle such as poor diet, obesity and lack of physical activity [11,12].
Over the last decade, most studies on the aetiology and development of metabolic diseases have indicated the increasingly important role of the gut microbiota. It is now known that dysbiosis of the microbiota is associated with significant metabolic consequences leading to abnormality of physiological processes in the body [13]. In fact, dysbiosis leads to increased permeability of the intestinal wall, resulting in elevated levels of bacterial endotoxins, (i.e., lipopolysaccharides - LPS) in the blood, which in turn can cause inflammation. Persistent inflammation can eventually lead to pathological metabolic conditions such as obesity, insulin resistance [14,15], metabolic syndrome [16,17], type 2 diabetes [18,19,20,21] and hyperglycaemia [22,23].
Selected microbial strains commonly known as probiotics have been documented to have a beneficial effect on glycaemic control in the blood (reviewed in [13,24,25,26]). In addition, there is growing evidence that microorganisms play an important role in glucose homeostasis, particularly in metabolic conditions such as obesity and obesity-induced insulin resistance [14,15], metabolic syndrome [16,17], type 2 diabetes [18,19,20,21,27] and cardiovascular disorders [23,28]. Despite different opinions on the terminology of probiotics [29], a microorganism can be classified as a probiotic if it meets most of the safety, functional, technological and physiological criteria [30,31]. Most probiotics derive from a heterogeneous group of lactic acid bacteria, dominated by bacteria from genus Lactobacillus [32], Lactococcus, Enterococcus and Streptococcus predominate [33]. Probiotics also include strains of other bacterial species, (i.e., Bifidobacterium sp. [34], Bacillus sp., Propionibacterium sp., Escherichia coli), yeasts (Saccharomyces) and moulds (Aspergillus) [33,35]. Although probiotics are “live microorganisms that when administered in adequate amounts confer a health benefit on the host” [29], their non-viable components named paraprobiotics may be a safer yet effective alternative for use in vulnerable individuals [36].
Despite the existence of several reviews mentioned above, knowledge of mechanistic studies on the beneficial effects of probiotics in glucose-related pathological conditions is scarce. Therefore, the aim of the review was to evaluate the most recent literature (between 2010 and 2021), in which probiotic administration has direct or indirect effects on glucose metabolism and its homeostasis. The review has been divided into sections regarding probiotic effects on the physiology of different human and animal organs. Concluding remarks and future perspectives on the importance of probiotics and their influence on glucose-related pathological disorders have been proposed.

2. Materials and Methods

Search Strategy and Studies’ Selection

Identification of the scientific literature accessible in English was performed using a combination of search terms (“probiotics; mechanisms; glucose”) in a publication period between January 2010 and December 2021. Studies were identified using the PubMed (https://pubmed.ncbi.nlm.nih.gov/, accessed on December 2021), ScienceDirect (https://www.sciencedirect.com/, accessed on December 2021) and Scopus (https://www.scopus.com/search/form.uri?display=basic#basic, accessed on December 2021) databases. The preliminary search yielded a total of 7079 records. Duplicates were excluded, as well as abstracts, due to insufficient available data. Further screening met the inclusion criteria, namely in vivo (human and animal) and in vitro studies showing any direct or indirect effect of probiotics (alone or in combination with other probiotics) on glucose, glucose metabolism or homeostasis. After considering all of the above criteria, 137 final studies were included in this review. The obtained data from the publications were further sorted based on the reported outcomes, (i.e., the different probiotic effects).

3. Effect of Probiotics on Glucose Metabolism and Homeostasis

The incomplete understanding of how probiotics work is due to a different methodological approach, a lack of investigation into mechanisms and major physiological differences between probiotic strains [37,38], which makes it difficult to comprehend the significance of their use, especially in glucose-related pathological conditions.

3.1. General Remarks about the Mechanisms of Probiotics

The qualitative and quantitative composition of the gut microbiota has a major impact on the interaction with the host. The current strategy for treating dysbiosis is still largely based on the use of probiotics to restore microbial diversity and normalise the “disturbed” gut microbiota [39]. Microbiota acts at different levels through complex mechanisms that are not fully understood [40]. As a result, studies of the potential mechanistic actions of probiotics often encounter a number of limitations:
  • The use of oversimplified in vitro models that often fail to reproduce the results in vivo;
  • The use of “human” probiotics in animal models in vivo, (e.g., rodents), which do not take into account the functional significance of the species- and strain-specific administration of probiotics and their impact on the host’s immune response and its microbiota;
  • Probiotics’ action ultimately requires the involvement of the endogenous microbiota, which is host-specific and often diet-dependent and therefore hardly reproducible;
  • Most of the mechanisms of probiotics presented in research studies have suggested two main principles of probiotics’ action [41];
  • Direct, contact-dependent principle (binding to different surface molecules);
  • Indirect principle via secretory molecules (production of bioactive peptides and metabolites).

3.2. Effect of Probiotics on Blood Parameters

3.2.1. In Vivo Human Studies

A number of human clinical trials demonstrated differential effects of probiotics on blood parameters in patients with various metabolic problems. These parameters reflect a direct or indirect relationship with glucose and its concentration in the blood (glycaemia) (Table 1).
Several studies have measured various blood parameters related to glycaemic status that may indicate possible control of blood glucose levels (glycaemic control). These can be divided into blood parameters, which are directly related to glycaemic status, such as fasting blood glucose, or blood parameters, which are indirectly related to glycaemic status, such as insulin, glycohaemoglobin (HbA1c) and homeostasis model assessment-estimated insulin resistance index (HOM-IR). However, some authors have included additional blood parameters, indirectly related to glycaemic status, in their studies, which are explained in more detail in Table 1. Tonucci et al. [50] have shown that glycaemic control is improved by taking probiotic fermented milk for 6 weeks. In one trial, administration of a probiotic preparation containing Lactobacillus casei Shirota [56] after 4 weeks maintained glycaemic control and preserved insulin levels in fasting state. A 6-month human clinical trial also reported beneficial effects of two probiotics (Lactobacillus reuteri ADR-1 and ADR-3) on patients with type 2 diabetes. ADR-3 reduced some parameters of glycaemic control, while ADR-1 had a beneficial effect on lowering blood pressure [51]. In another 12-week human clinical trial, probiotic did not alter HbA1c levels, fat levels or total serum bile acids [54]. Furthermore, Shellekens et al. [45] have demonstrated some anti-obesity effects of Bifidobacterium longum APC1472 in humans, possibly due to an alternation in ghrelin signalling. Since the gut hormone ghrelin may be involved in glucose homeostasis via inhibition of insulin secretion [57], administration of Bifidobacterium longum APC1472 may be clinically significant in patients with pre-diabetes and type 2 diabetes mellitus. In addition, the authors speculated that the reduction in fasting cortisol levels due to probiotic administration affects the hypothalamic–pituitary–adrenal (HPA) axis, which regulates the pancreas and simultaneously influences glucagon and insulin secretion. All of these effects may contribute to glucose level reduction. A 12-week administration of Lactobacillus plantarum OLL2712 in pre-diabetic patients has shown improvement in fasting plasma glucose levels, glycoalbumin levels and insulin resistance [46]. In a trial in healthy adults, administration of the paraprobiotic Lacticaseibacillus casei 01 resulted in a better reduction in blood glucose levels than the probiotic of the same name [42].
The HOM-IR index is an important parameter in the context of cardiological and metabolic disorders, as it contributes to the early detection of insulin resistance and to the assessment of the risk of developing diabetes, cardiovascular pathologies and atherosclerosis [58]. The effect of probiotics on blood parameters was also demonstrated in a clinical trial conducted in the Arab population [47], in which a 6-month intake of a multi-strain probiotic supplement (Ecologic® Barrier) showed a decrease in the HOM-IR index as well as a decrease in the level of endotoxin and inflammatory adipokines. The authors suggested the use of the probiotic supplement as a dietary supplement for patients with type 2 diabetes, but also emphasised the wide variation in results due to genetic diversity, dietary habits and environmental differences (geographical regions) between the populations studied [47]. Kobyliak et al. [52] also demonstrated the impact of a multi-strain probiotic (“Symbiter”) on reducing HOM-IR. Similar results were reported by Khalili et al. with Lactobacillus casei 01 [49] and Lactobacillus casei [48] as a supplement. In patients with metabolic syndrome, a two-month intake of probiotic yoghurt (Lactobacillus acidophilus La5 and Bifidobacterium lactis Bb12) led to a reduction in blood glucose levels and significant changes in insulin resistance (HOMA-IR) and sensitivity. The authors therefore believe that regular consumption of probiotic yoghurt has a positive effect on the treatment of metabolic syndrome [44].
Interleukin-6 (IL-6), together with monocytochemotactic protein-1 (MCP-1), is a pro-inflammatory cytokine associated with the development of insulin resistance and hyperglycaemia [52]. In addition, a pro-inflammatory interleukin 1 beta (IL-1β) cytokine may be involved in postprandial inflammation and thus in the regulation of glucose homeostasis and immune response [59], while tumour necrosis factor alpha (TNF-α) seems to be mainly involved in insulin signalling and action [60] and in the regulation of glucose and lipid metabolism [61]. However, Kobylak et al. additionally showed that glycaemia-related parameters such as body weight, body mass index (BMI) and IL-6 remained unchanged or did not significantly decrease (HbA1c) after administration of multi-strain probiotics. However, the same compounded probiotics altered some pro-inflammatory factors such as TNF-α and IL-1β [52]. Toshimitsu et al. [46] have shown that both parameters as well as IL-6 were suppressed after 12 weeks of treatment with Lactobacillus plantarum OLL2712 in a pilot trial with pre-diabetic patients.
HbA1c is an important parameter in the long-term monitoring of blood glucose levels (balance) and indicates the number of glucose molecules bound to haemoglobin in the red blood cells (erythrocytes) [62]. The mechanism by which probiotics reduce the amount of this parameter is still unclear [26]. Five clinical studies [43,50,51,52,53] showed a statistically significant reduction in HbA1c levels after taking a probiotic preparation, while two other studies found no change in the amount of this parameter [49,63].
Fetuin-A is known as a glycoprotein, which is secreted by liver and adipose tissue. Its increase in the blood is indirectly related to patients with atherosclerosis, insulin resistance, diabetes mellitus and metabolic syndrome [64], as it affects the activity of insulin receptor tyrosine kinase [65]. Sirtuins (SIRTs) are very important regulators of energy homeostasis and metabolism due to their deacetylation activity in variety of organs [66]. Fetuin-A and SIRTs were investigated in a randomised controlled trial in patients with type 2 diabetes after an 8-week intake of Lactobacillus casei 01 and Lactobacillus casei [48,49]. The results of blood analysis showed that fetuin-A levels were decreased, while SIRTs levels were increased.
Despite the proven beneficial effects of probiotic therapy in patients with type 2 diabetes, some studies failed to demonstrate changes in blood and metabolic parameters or the changes were not statistically significant, which is a crucial justification for a more objective approach and evaluation of the effectiveness of probiotic therapy. The diversity of results is mostly a consequence of the different methodological approaches in the studies: use of one or more probiotic strains, their amount and duration of administration and the number of patients. Moreover, it is already known that the activity of probiotics is species-specific [67]. A comparison of the different results of the individual clinical probiotic studies can be carried out within the framework of meta-analytical studies. In this way, a more objective view of the potentially beneficial effect of probiotics on various blood and metabolic parameters in patients with type 2 diabetes can be obtained [67,68,69,70,71,72,73,74,75,76]. However, the above meta-analyses point to numerous limitations, even if they ultimately confirm the multiple beneficial effects of probiotics on blood and metabolic parameters: small groups of study participants [67,68,69,71,74], lack of data on dietary habits and physical activity of study participants [68,70,73], diversity within a study [69,71,72,73,74], short duration of probiotic therapy, different therapeutic doses, use of different probiotic strains [67,68,70,72,74], lack of evidence on possible side-effects of probiotic use [69] and lack of testing of other metabolic factors affecting glycaemic control [70,71]. Last, but not least, Tao et al. [76] proposed that more clinical data and research on the mechanisms of probiotic are needed.

3.2.2. In Vivo Animal Models Studies

Many studies in animal models demonstrated positive effects of various probiotics on diverse blood parameters, which are summarised in Table 2. The range of measured parameters was similar to in vivo human studies showing potential effects of probiotics on improving glucose tolerance, insulin resistance and insulin sensitivity.
Oral intake of probiotic preparations of the strains Lactobacillus casei and Bifidobacterium bifidum (alone or both simultaneously) reduced the incidence of hyperglycaemia (increased blood glucose concentration) and dyslipidaemia (imbalance of blood lipid concentration) in rats with induced diabetes [79]. Similar antidiabetic effects in rats were obtained by administration of probiotic strains Lactobacillus plantarum TN627 [80], Bifidobacterium animalis 01 [81], Lactobacillus paracasei HII01 [82], 9 strains of Lactobacillus rhamnosus, Bifidobacterium adolescentis and Bifidobacterium bifidum [77], Lactobacillus rhamnosus BSL and R23 [83], heat-inactivated Streptococcus thermophilus [84], Lactiplantibacillus plantarum IMC 510 [85] and Lactobacillus fermentum RS-2 [86]. In the latter case, in addition to reduced glucose levels in fasting state, a low oxidative stress was observed [86].
Studies in mice and rats have shown, among other things, a positive effect of probiotic preparations on glucose balance [78,87,88,89,90,91,92,93,94,95,96,112,113,118] and reduced insulin resistance [78,87,89,90,91,92,94,113]. Andersson et al. [97], Sakai et al. [98], Park et al. [114], Naito et al. [99], Wang et al. [100,101], Machado et al. [102], Lee et al. [103], Hsu et al. [115], Kim et al. [116] and Zeng et al. [104] have shown that the probiotic addition of different species of the genus Lactobacillus or a combination of probiotics [100,101,116] improves glucose sensitivity (tolerance) [97,98,99,100,101,102,103,114,115,116] and reduces insulin resistance [97,98,99,100,101,114] by decreasing serum glucose levels in obese/diabetic mice and rat models. Endotoxaemia, along with inflammation, is a common factor in metabolic diseases (especially obesity, insulin resistance and diabetes). In a study conducted in mice, the addition of Lactobacillus casei Shirota reduced insulin resistance and endotoxaemia. In this context, it has been demonstrated that the probiotic effect on glucose regulation is not necessarily directly related to the body weight and the amount of fat in the animals [99].
Effects on glucose homeostasis and cholesterol metabolism have also been demonstrated by Ashrafian et al. [105], Wang et al. [106], Kim et al. [116], Tarrah et al. [107] and Sun et al. [108]. In the latter study in mice, Lactobacillus acidophilus SJLH001 had a statistically significant effect on the regulation of transcription genes, essential for glucose transport and cholesterol metabolism [108]. Yan et al. [110] have demonstrated a beneficial effect of Lactobacillus acidophilus on type 2 diabetes in mice by controlling liver glucose levels, lipid metabolism and gut microbiota.
Furthermore, in a study on mice [78,91,94,113,117] and rats [84,89], serum levels of certain inflammatory markers such as IL-6, TNF-α and IL-1β were decreased after a specific probiotic administration. In addition, heat-killed Streptococcus thermophilus [84], Bifidobacterium animalis 01 [81] and Lactobacillus casei LC89 [91], Lactobacillus plantarum CCFM0236 [78] increased the levels of IL-10, an important anti-inflammatory marker, when administered to rats and mice with type 2 diabetes, respectively.
A study in rats [81] showed a statistically significant reduction in HbA1c levels after taking probiotic supplements. In addition to ageing and neurodegenerative diseases, impaired memory function in the brain may also be associated with impaired glucose metabolism, i.e., an increase in plasma HbA1c levels [117]. Moreover, Bonfili et al. have demonstrated the beneficial effects of the probiotic formulation SLAB51 in a mouse model of Alzheimer’s disease. The probiotic formulation lowered serum HbA1c levels, showing a positive effect on glucose homeostasis [111].
Osteocalcin is a protein hormone produced by osteoblasts. Primarily it inhibits bone formation, but as a hormone it plays an important role in the synthesis of testosterone in testis as well as synthesis of the muscle mass [119]. However, it has been proven to have a positive effect on improving glucose tolerance and regulating glucose metabolism [119,120,121]. The addition of Lactobacillus casei Zhang in hyperinsulinaemic rats increased the amount of osteocalcin, which in turn influenced the improvement of glucose tolerance. The regulation of glucose tolerance is probably induced via the osteocalcin–adiponectin pathway [109].

3.2.3. In Vitro Studies

The enzyme alpha-glucosidase (α-glucosidase) catalyses the digestive processes of carbohydrates and is responsible for increased blood glucose levels [118]. In in vitro studies [78,94,108,118,122], certain probiotics inhibited α-glucosidase activity and thereby decreasing the level of glucose ready for absorption and thereby glycaemic index of food.

3.3. Effect of Probiotics on Brain Function

The apparent communication between the intestinal microbiota and the central nervous system via the “gut-brain axis” has led researchers and/or clinicians to investigate the preventive and therapeutic use of probiotics in various neurological disorders [123].
Recent studies have revealed an important role of glucose homeostasis in some brain pathological conditions, such as Alzheimer’s disease (AD) [117]. Bonfili et al. have shown that the administration of probiotic SLAB51 increased glucose uptake in mice with Alzheimer’s disease due to the restoration of glucose receptor 3 (GLUT3) and glucose receptor 1 (GLUT1), the key glucose receptors in brain. The increased expression of both glucose receptors was in line with alleviated levels of phosphorylated 5′ adenosine monophosphate-activated protein kinase (AMPK) and protein kinase B (AKT), which are important metabolic regulators in most cell types. Moreover, they reported an increased expression of brain insulin-like growth factor receptor β (IGF-IRβ). IGF-IRβ is considered as a promising therapeutic target in AD, because of its important part in proteasome-mediated removal of oxidised proteins [111].

3.4. Effect of Probiotics on Bile Acid Metabolism

Bile acids are highly effective surface-active digestive substances (surfactants) and natural emulsifiers whose main function is to support the digestion and absorption of fat-soluble lipids and vitamins. Their cholesterol production takes place exclusively in the hepatocytes. After they have fulfilled this function in the small intestine, they are reabsorbed into the bloodstream as part of the so-called “enterohepatic recirculation”. From here, their path leads back to the liver, where their activity is related to the various signalling pathways [124].
Probiotics containing bile salt hydrolase (BSH) have been shown to interfere with the metabolism of bile acids and promote the deconjugation of bile salts via BSH, which is important in chronic metabolic diseases. In human studies, daily addition of Lactobacillus reuteri NCIMB 30242 increased the amount of free bile acids that were no longer absorbed in the intestinal wall as a result of probiotic BSH deconjugation, but were excreted in the faeces. It was also speculated that deconjugation might be directly related to the increase in fibroblast growth factor 19 (FGF) [125]. In fact, FGF plays an important role in the regulation of bile acid synthesis and its amount directly influences lipid and glucose metabolism [126].
In a human clinical study, Lactobacillus reuteri DSM 17938 can contribute to an increase in the level of deconjugated bile acids, which was also evidenced by the positive correlation with an increase in insulin sensitivity and thus an improvement in glucose metabolism. The authors hypothesised that the probiotic influences the biosynthesis of deconjugated bile acids by regulating genes for certain crucial enzymes involved in the biosynthesis [54].
In a study in rats the probiotic Lactobacillus casei Zhang [109] increased gene expression of liver X-receptor-α (LXR), which has already been shown to increase glucose tolerance and cholesterol reduction via increased BSH levels and deconjugation of bile acids [127]. In another study conducted in rats [128], the same probiotic influenced the reduction of bacteria with hydrolase activity, resulting in increased bile acid secretion.

3.5. Effect of Probiotics on Adipose Tissue Function and Inflammation

For many years, adipose tissue was considered to be an inactive organ with a single function, the storage of lipids. However, a number of studies have shown that it is an active organ, which plays an essential metabolic and hormonal role in homeostasis of energy metabolism and glycaemic regulation [16,129] (Figure 1). Excessive accumulation of lipids in the adipose tissue leads to its hypertrophy, cellular stress and local inflammation. The latter is chronically associated with obesity, insulin resistance, hyperglycaemia and type 2 diabetes [130].
In studies on high-fat diet fed mice, some probiotic strains have demonstrated their protective role against chronic inflammation in adipose tissue, leading either to its prevention or its inhibition by reducing insulin resistance and hyperglycaemic state. Indeed, a high-fat diet has been shown to increase some of the pro-inflammatory factors such as fat TNF-α, IL-6, IL-1β. Lactobacillus fermentum MTC5689 decreased the expression of pro-inflammatory indicator genes, TNF-α and IL-6 in visceral adipose tissue of mice [131]. In the second study in mice, Lactobacillus sakei OC67 reduced the incidence of hyperglycaemia and obesity by lowering inflammation [132]. In both studies, as well as in three other studies [98,133,134] reductions in glucose levels in fasting state and a decrease in IL-1β expression have been reported. The reduction of TNF-α expression in fat has also been shown after the addition of Lactobacillus fermentum LM1016 [133], Lactobacillus crustorum MN047 and Lactobacillus rhamnosus LS-8 [134]. The later probiotic decreased the IL-6 expression in adipose tissue as well [134]. The effect of Lactobacillus rhamnosus GG also showed the improvement and reduction of glucose hypersensitivity and insulin resistance in obese mice. The addition of the probiotic was able to reduce the expression of genes for specific macrophage factors important for macrophage infiltration and activation, leading to a reduction in macrophage activation, inflammation and thus improved insulin sensitivity in adipose tissue [114].
Deng et al. [135] suggested that different genotypes of Akkermansia municiphila exert specific effects in repairing damage to adipose tissue caused by a high-fat diet in mice. The study showed that different genotypes of probiotic can play different roles in the same disease state, particularly in combating endotoxaemia, inflammation and whitening of brown adipose tissue, glucose tolerance, hyperlipidaemia and hepatic steatosis.
Strains of the genera Bifidobacterium [136] and Bacillus [113] have been shown to influence the increased expression of proteins involved in the insulin signalling pathway in mice. Adiponectin, along with leptins, are involved in controlling feeding behaviour and increasing insulin sensitivity [137]. Increased adiponectin mRNA levels in fat cells (adipocytes) [136] and increased serum leptin levels [113] have also been demonstrated in the above studies. Due to the increased insulin sensitivity, probiotics influenced the improvement of glucose absorption in tissues by elevating adiponectin and leptin concentrations, which led to a decrease in blood glucose levels [138]. However, in a study in mice [139], adiponectin blood levels as well as its mRNA levels in adipose tissue were decreased and restored by the administration of probiotic supernatants.
Peroxisome proliferator-activated receptor α (PPARα) is important for processes controlling energy expenditure, inflammation and glucose homeostasis [140]. Due to increased levels of a hepatokine mediator of adiponectin expression in the regulation of glucose and lipid metabolism-fibroblast growth factor 21 (FGF21), increased PPARα mRNA levels and butyric acid levels, the authors suggested a possible link between probiotic mechanism-butyric acid-PPARα-FGF21-adiponectin-glucose and lipid metabolism [139]. Indeed, Molina-Tijeras et al. [141] linked the improvement in glucose and lipid metabolism to the upregulation of PPARα expression after administration of Lactobacillus fermentum CECT5716.
The downregulation of sirt1 (sirtuin-silent mating type information regulation 2 homolog 1) gene expression is an indicator of metabolic dysregulation in many organs, including adipose tissue. In a study on mice, Lactobacillus plantarum increased adiponectin levels, which may have led to an upregulation of sirt1 gene expression and thus to an improvement in lipid metabolic dysfunction [142].
The role of the major adiponectin receptor gene, i.e., adiponectin receptor 2 (AdipoR2), has been linked to insulin resistance and type 2 diabetes, where it plays a crucial role in glucose and lipid metabolism, inflammation and oxidative stress [143]. In a study on rats [109], the probiotic Lactobacillus casei Zhang also increased the expression of AdipoR2 in fat cells. The same probiotic has also increased the expression of the genes for the glucose receptor 4 (GLUT4) and core receptor-peroxisome proliferator-activated receptor gamma (PPAR-γ). Both parameters consequently had a positive effect on oral glucose tolerance. PPAR-γ is a known receptor that maintains the expression of key glucose regulatory and liporegulatory molecules, important for insulin signal transduction [144], while GLUT4 is the major glucose transporter for the facilitated transfer of glucose from blood to skeletal fat tissue and muscle [145]. A possible link between improved glycaemic levels, utilisation of glucose and increased expression of GLUT4 genes following administration of Lactobacillus fermentum CECT5716 has also been proposed [141]. In addition to PPAR-γ, some probiotics have also been shown to affect increased expression of PPAR-α. in animal models [105,146] and in one in vitro study [147]
A transforming growth factor beta (TGF-β) is known to be an important parameter in adipose tissue due to its effects on inflammatory mediators, energy homeostasis, fat expansion and collagen deposition [148,149]. A decrease in TGF-β mRNA levels after administration of Akkermansia muciniphila as well as its extracellular vesicles (EVs) suggested a potential antidiabetic and anti-obesity effect of both the bacterium and its EVs [105].
Lactobacillus plantarum Ln4 significantly reduced lipid deposition and induced glucose absorption in 3T3-L1 adipocytes in an in vitro study [150].

3.6. Effect of Probiotics on Skeletal Muscle

The endoplasmic reticulum (ER) plays a role as a sensor for changes in the homeostasis of a cell function. Stress therefore disrupts the normal function of the ER, which in turn reacts differently to protect the cell. It is known that lipid metabolism and ER stress are closely linked in skeletal muscle cells. In addition, ER stress in muscle cells is also a major cause of insulin resistance, which in turn affects glucose homeostasis [151]. In a model study in mice, certain strains of Lactobacillus species have been shown to influence the reduction of ER stressors in skeletal muscle, thereby reducing lipotoxicity. In fact, the probiotic effect showed a decrease in the expression of certain genes that can alter lipid metabolism [114,131].
Administration of Lactobacillus casei or Bifidobacterium bifidum strains or a combination of both significantly increased the amount of muscle glycogen in rats with induced diabetes, which had a direct effect on reducing hyperglycaemia [79]. Similarly, Liu et al. showed an increase in protein levels of some glucose homeostasis-related molecules such as phosphoinositide 3-kinase (PI3K), phosphorylated protein kinase B (p-AKT) and glycogen synthase kinase 3 beta (GSK-3β). The latter is directly related to increased glycogen production [139].
The protective effect of Lactobacillus plantarum was demonstrated in a study conducted on mice [142]. The expression of irisin, a specific adipomyokine produced by skeletal muscle and adipose tissue, was upregulated, which in turn increased the insulin sensitivity of skeletal muscle and improved hepatic glucose and lipid metabolism. Since irisin is a peroxisome proliferator-activated receptor-γ-coactivator (PGC)-1α-dependent molecule, the authors speculated on a possible concurrent probiotic mechanism for improving metabolism based on the regulation of mitochondrial biogenesis.
Kim et al. [152] have also demonstrated the effect of a probiotic on increased expression of genes relevant to glycogen synthesis (glycogen synthesis-related gene pp-1) and increased expression of GLUT4 in skeletal muscle. In the same in vitro study, the authors showed improved insulin-dependent glucose uptake and increased expression of the GLUT4 and PPAR-γ genes.
The increase in GLUT4 expression due to the effect of the probiotic additive has been demonstrated in three animal studies: in mice [153] and rats [82,146]. Toejing et al. [82] linked the activation of GLUT4 translocation to the membrane in skeletal muscles to AMPK activation, which consequently affects the glucose uptake. The effects of probiotics on skeletal muscle in relation to glucose homeostasis and metabolism are shown in Figure 2.

3.7. Effect of Probiotics on Liver, Pancreas and Kidney

A pathological state of diabetes reduces the antioxidant potential of pancreatic and hepatic tissue and consequently increases the harmful effects of free radicals [154]. Studies on animal models have shown that single probiotics reduce oxidative stress in the pancreas [79,104,155], liver [90,93,104,156] and kidney [93,104], thus helping to increase the antioxidant activity and physiological function of the organ.

3.7.1. Liver

Figure 3 shows some crucial effects of selected probiotics on the liver that lead to a change in glucose metabolism.
Increased liver glycogen levels and decreased blood glucose levels after supplementation with Bacillus toyonensis SAU-19 in high-fat diet/streptozicin-induced diabetic mice indicated a probiotic effect on glycogen synthesis [90]. Some parameters of liver metabolism, including liver glucose decreased after administration of Lactobacillus plantarum wild type strain (δplnEFI) in a study on diet-induced obese mice [157].
The addition of Bifidobacterium lactis HY8101 [156] and Lactobacillus fermentum LM1016 [133] to mice decreased the expression of genes relevant to the regulation of hepatic gluconeogenesis, i.e., phosphoenolpyruvate carboxykinase 1 (PCK1) and glucose-6-phosphatase catalytic subunit (G6PC), resulting in a reduction of blood glucose levels in fasting state. The same parameters, as well as β-cell analysis, were studied after administration of Lactobacillus plantarum HAC01 [103] and a multi-strain probiotic supplement ProbiogluTM [89] in diabetic mice. In both studies, the probiotics had a protective and restorative effect on β-cells. Improvement in glucose tolerance and insulin sensitivity due to the downregulation of G6PC [83,158] and phosphoenolpyruvate carboxykinase (PEPCK) [158] was also reported in diabetic rats by Farida et al. [83] and by Huang et al. [158] in olanzapine-induced diabetic mice, when they administered Lactobacillus rhamnosus BSL/Lactobacillus rhamnosus R23 and Akkermansia muciniphila, respectively. For both parameters, similar results were also observed by Okyere et al. after administration of Bacillus toyonensis SAU-19 [90]. Moreover, an improvement in glucose metabolism has been demonstrated by the upregulation of genes related to glycogenesis and glucose transport, such as Forehead Box O1 (FOXO1), glycogen synthase (GS), phosphofructokinase-1 (PFK-1) and glucose transporter 2 (GLUT2), respectively.
Park et al. [159] decreased gene expression of the enzyme PEPCK, which is critical for hepatic gluconeogenesis, by supplementing high-fat diet-fed mice with Lactobacillus acidophilus NS1 (NS1). The reduced expression of the enzyme was also confirmed in vitro by demonstrating that NS1 regulates reduction by activating transcription of hepatocyte nuclear factor 4 alpha (HNF4α), a core receptor and a key element in hepatic differentiation and cell proliferation [160].
Glucagon is an important hormone for maintaining blood glucose homeostasis. It regulates the activation of certain molecules in its pathway, such as the heterotrimeric Gs protein alpha-subunit (Gnas), cAmp-dependent protein kinase (PKA), CREB-regulated transcriptional coactivator 2 (CRTC2), as well as PEPCK and G6Pase [161,162]. Zhang et al. have demonstrated a probiotic effect on the glucagon signalling pathway in the liver. Lactobacillus casei LC89 downregulated all of the above molecules in the glucagon signalling pathway. Moreover, a reduction in blood glucose and an increase in liver glycogen have been demonstrated. Both parameters are also closely related to the downregulated genes of the above molecules [91].
The renin-angiotensin system (RAS) is a well-defined regulator of blood volume (its pressure) and total vascular resistance [163]. One of the major components of RAS is the angiotensin-converting enzyme 2/angiotensin-(1–7)/mitochondrial assembly receptor (ACE2/Ang-(1–7)/MasR) axis. Once activated, this axis plays a crucial role in many organs, especially in the liver, where it has already shown its potential as a regulator of many metabolic disorders [164,165]. Machado et al. [102] has demonstrated the potential effect of Bifidobacterium longum administration on improving metabolism in the liver of obese mice by increasing the expression of hepatic ACE2 and MASR. The increased expression of both parameters is directly related to the increase in Ang-(1–7), an important vasodilating agent that can reduce gluconeogenesis in the liver and improve glucose and lipid metabolism [166].
In two studies conducted in mice, Lactobacillus paracasei TD062 and Lactobacillus plantarum HAC01 also reduced the expression of certain glucose-linked genes (PEPCK) and had a positive effect on the PI3K/AKT signalling pathway, which is crucial for the biological activity of insulin and glucose uptake [103,122]. A similar effect was observed in a rat study [81] after administration of Bifidobacterium animalis 01. However, Wang et al. [100] have demonstrated a putative link between the protective effect of probiotics on pancreatic apoptosis and the upregulation of the PI3K/AKT pathway. In addition, probiotic administration showed protective properties against oxidative damage by affecting Kelch-like ECH-associated protein 1/nuclear factor-erythroid factor 2-related factor 2 (Keap1/Nrf2). The latter is important for the regulation of antioxidant genes [81]. The downregulation of Nrf2 expression associated with oxidative stress in the liver was also demonstrated by Wang et al. when they administered Bacillus amyloliquefaciens SC06 to mice fed a high-fat diet [113]. A positive effect on insulin sensitivity was also observed in mice after administration of Lactobacillus rhamnosus GG supernatant [139] and two strains of Lactobacillus acidophilus [110] with significant increases in PI3K [139] and phosphorylated (p)-AKT [110,139] as well as GSK-3β [110,139].
In high-fat diet and streptozoticin-induced diabetes mice, five different probiotic strains out of nine tested [77], two different Lactobacillus acidophilus strains [110] and Bacillus toyonensis SAU-19 [90] reduced TNF-α [77,90,110], IL-1β [77,90,110] and IL-6 [77,110] in liver homogenates pointing out their anti-inflammatory effect, which is also consistent with their hypoglycaemic effect, i.e., reduction in serum glucose levels [77,90].
Three probiotic strains, Lactobacillus paracasei 1F-20, Lactobacillus fermentum F40-4 and Bifidobacterium animalis subsp. lactis F1-7, were selected to test their effect on glucose uptake in HepG2 cells (hepatocytes), leading to their enhancement and thus promoting glucose metabolism in the liver [147].
However, the efficacy and safety of the use of probiotics in liver is still the subject of research, which is why Stavropoulou and Bezirtzoglou [123] do not yet recommend their use for the treatment of most liver diseases.

3.7.2. Pancreas

Some in vitro studies and studies on experimental animals have shown the effect of probiotics on preventing damage to pancreatic tissue by preventing degeneration and inflammation of pancreatic islets or inducing their regeneration. Therefore, the addition of probiotics is expected to improve glucose tolerance [77,78,91,96,118,156,167]. In addition, Wang et al. [100,101] have shown a significant improvement in pancreatic morphology and insulin secretion as a result of probiotic administration. In a study with Goto-Kakizaki rats, Lactobacillus gasseri SBT2055 increased insulin secretion via increased expression of insulin genes (Ins1 and Ins2) and an increased transcription factor for insulin genes—pancreatic and duodenal homeobox (PDX1)—showing improvement in pancreatic inflammatory status and a decrease in serum glucose levels [168].
Hyperglycaemia is also associated with loss of chloride ions. High intracellular concentrations of chloride ions in pancreatic β-cells are critical for the electrical activity of the β-cell membrane and insulin secretion. The addition of the Lactobacillus casei Zhang strain in a rat study increased of chloride channel protein 2 (ClC-2) expression in chloride channels, resulting in reduced chloride ion loss and improved electrical activity of the β-cell membrane and insulin secretion [131].

3.7.3. Kidney

Insulin receptor substrate-1 protein (IRS-1), AKT and endothelial nitric oxide synthase (eNOS) are important components of glucose homeostasis signalling pathways and lipid metabolism in the kidney. In addition, sodium-glucose cotransporter 2 (SGLT2) is the major glucose transporter, while glucose transporter 5 (GLUT5) is the specific fructose transporter [145,169]. In a study on rats fed a high fructose concentration, Korkmaz et al. [170] demonstrated a positive effect of Lactobacillus plantarum and Lactobacillus helveticus strains on the insulin signalling pathway, inflammatory markers and renal glucose transporters. Lactobacillus plantarum and partially Lactobacillus helveticus enhanced the expression of IRS -1, AKT and eNOS. In addition, Lactobacillus plantarum decreased the level of individual inflammatory factors and the expression of the IL-6 and SGLT2 genes, with unchanged expression of the GLUT5 gene. Various physiological changes caused by the administration of selected probiotics in the pancreas and kidney are shown in Figure 4.

3.8. Effect of Probiotics on the Intestine

After ingestion, the intestine is the target organ of the probiotic and therefore of utmost importance for its various mechanisms of action. Figure 5 shows some common effects of probiotics in the intestine and the impact of their action on glucose metabolism and homeostasis.

3.8.1. Effect of Probiotics on the Intestinal Microbiota

A typical intestinal microbiota of an adult consists primarily of a set of six or seven different phyla, of which bacterial species of the phyla Bacteroidetes (Bacteroidota) and Firmicutes (Bacillota) predominate [171]. Bacterial species of the phyla Proteobacteria (Pseudomonadota), Verrucomicrobia (Verrucomicrobiota), Actinobacteria (Actinomycetota) and Euryarchaeota make up a smaller proportion [172]. Dysbiosis is often associated with various pathological conditions, including metabolic diseases such as obesity and type 2 diabetes [13]. In both diseases, the bacteria Akkermansia muciniphila and Faecalibacterium prausnitzii show a negative correlation [34]. Furthermore, Papadopolous et al. [28] emphasise in their review that dysbiosis of the gut microbiota is the decisive cause for a large number of risk factors for cardiovascular diseases.
Over the last decade, studies in mice have clearly shown a link between the gut microbiota and obesity. The ratio between the number/concentration of the phyla Firmicutes (Bacillota) and Bacteroidetes (Bacteroidota) has shifted in favour of the phylum Firmicutes (Bacillota) in obese mice and mice fed with high-calorie diet [113,173,174]. In addition, bacteria of the phylum Bacteroidetes (Bacteroidota) have fewer genes encoding enzymes relevant for the degradation of lipids and carbohydrates [175]. However, the results of previous studies are very different, even contradictory [123]. They show a positive [176,177,178,179] or negative [180,181] relationship between bacteria of the phyla Firmicutes (Bacillota)/Bacteriodetes (Bacteroidota) and the body mass index (BMI) or statistically non-significant relationships [182,183]. With regard to the higher proportion of Lactobacillus bacteria and the lower proportion of Bifidobacterium bacteria in obese people, the literature is consistent [184]. In addition, a decrease in metabolic values as well as weight gain after taking probiotics was observed in obese adults [123].
Although type 2 diabetes can be considered a pathological condition associated with obesity, metagenomic studies reveal a typical structure of the intestinal microbiota in patients with type 2 diabetes [24,34]. In addition, Larsen et al. [185] have demonstrated a positive correlation between the phyla Bacteroidetes (Bacteroidota)/Firmicutes (Bacillota) ratio and serum glucose levels. However, it has been shown that the microbiota of these individuals often contains a lower proportion of butyric acid producing bacteria and thus several opportunistic bacteria. Furthermore, by analysing the structure of the gut microbiota of patients with type 2 diabetes or those prone to developing the disease, physicians can improve the prognosis or course of the disease by preventing a deterioration in glycaemic status, and thus, the development of insulin resistance [13]. However, it is recommended to include functional foods with probiotic properties in the daily diet of patients with type 2 diabetes to improve metabolic imbalance and control and prevent the severity of the disease [27,123].
  • In vivo human studies
In a human study, Ivey et al. [186] showed no effect of consuming probiotic yoghurt and probiotic capsules on the composition of the microbiota. They reasoned that the performance and efficacy of probiotic bacteria depend on the specific conditions in the gut. Indeed, the expression of probiotic genes is influenced by both the genotype of the host and the genotype of the interacting gut bacteria. In addition, the host’s diet can also influence the metabolism of probiotics [186]. In another human study, no change in the composition of the gut microbiota was observed after probiotic addition, which was explained by the fact that the probiotic metabolic response depends on the initial composition of the host gut microbiota, i.e., before the start of therapy [54]. An eight-week randomised control trial in children and adolescents with obesity showed a change in microbiota (reduced number of Escherichia coli), weight and an improvement in insulin sensitivity and certain metabolic parameters after supplementation with Bifidobacterium breve BR03 and B632 [187]. In patients with type 2 diabetes, the use of different Lactobacillus reuteri ADR-1/3 strains affected the modulation of the intestinal microbiota (increase in fecal Lactobacillus reuteri levels), resulting in HbA1c reduction and alleviation of type 2 diabetes symptoms [51].
Early pregnancy is generally characterised by glucose and insulin levels being within the normal range [188]. However, in some cases, an increase in serum insulin levels is observed in late pregnancy, often associated with insulin resistance, which can lead to glucose hypersensitivity and type 2 diabetes—gestational diabetes mellitus (GDM) [189]. Insulin resistance is thus the cause of inflammation (increase in various inflammatory factors) and depends on the structure of the intestinal microbiota, which has a significant influence on insulin metabolism. GDM not only has negative effects on the mother, but also on the newborn, who shows signs of macrosomia and hypoglycaemia [190]. Systematic studies [190] on the use of probiotics (mostly Lactobacillus and Bifidobacterium) indicate their beneficial effect on GDM in the period between pregnancies. Probiotics can alter control over the gut microbiota [189], helping to modulate the immune system, reduce inflammatory factors and improve glucose and blood sugar control [189,191,192,193]. Ebrahimi et al. [194] also showed a positive effect of probiotic yoghurt consumption on blood glucose control, i.e., a significant decrease in blood glucose and HbA1c levels in pregnant women. On the other hand, Shahriari et al. [195] showed neither a significant reduction in the risk of GDM nor an improvement in other neonatal and maternal outcomes after oral administration of a probiotic supplement mixture.
The exact effects and thus the relationship between probiotics and the intestinal microbiota are still not fully understood, indicating some conflicting studies. It seems that the relationship between the intestinal microbiota and probiotics is unique to each individual and should be studied as such [196].
  • In vivo animal model studies
In the studies listed below, changes in the Bacteroidetes (Bacteroidota)/Firmicutes (Bacillota) ratio and in the relative abundance of the two phyla after different probiotic administrations may represent the best parameter for evaluating the beneficial effects of probiotics on the gut microbiota. Restoration of the two phyla was reported regardless of the pathological conditions of the animal. Since it is already known that there is a positive correlation between the ratio/relative abundance of the two phyla and glucose in some pathological conditions [192], it is not surprising that the administration of certain probiotics could improve or restore glucose tolerance and insulin sensitivity, possibly through direct or indirect modulation of the host gut microbiota. This includes increasing the relative abundance of Bacteroidetes (Bacteroidota) and decreasing that of Firmicutes (Bacillota), resulting in a decrease in the Firmicutes (Bacillota)/Bacteroidetes (Bacteroidota) ratio. Table 3 summarises some modulatory effects of selected probiotics administration on the microbiota in relation to glucose metabolism and homeostasis in animal model studies.
Zhang et al. [91] showed a positive correlation between phyla Bacteroidetes (Bacteroidota)/Firmicutes (Bacillota) ratio and serum glucose levels after administration of Lactobacillus casei LC89 to diabetic mice. They even suggested that the reduction in Firmicutes (Bacillota) and increase in Bacteroidetes (Bacteroidota) could be an additional factor in improving glucose tolerance. After 11 weeks of treatment with Lactobacillus plantarum HT121 [197] and Q180 [146], a reduction in body weight and an improvement in glucose tolerance were observed in mice fed with a high-fat diet, which could correlate with the restored Firmicutes (Bacillota)/Bacteroidetes (especially Akkermansia, Lactobacillus and Ruminococcus [197]) ratio. Li et al. [198] showed in rats that administration of the probiotic Lactobacillus casei CCFM419 increased the proportion of bacteria belonging to genus Allobaculum and Bacteroides. These changes may help to alleviate type 2 diabetes. In another study in diabetic mice, changes in the intestinal microbiota were observed after treatment with Lactobacillus plantarum HAC01, i.e., an increase in the Verrucomicrobia (Verrucomicrobiota) phylum (Akkermansia muciniphila) and a decrease in the Proteobacteria (Pseudomonadota) phylum (Dusulfovibrionaceae family). The increased levels of Akkermansia may have contributed to increasing insulin sensitivity [103]. Furthermore, in a study with diabetic mice [101], 14 compound probiotics reduced the number of harmful bacteria such as Bacteroides thetaiotaomicron, whose polysaccharose degradation leads to enhanced glucose and energy absorption, resulting in hyperglycaemia. Li et al. [199] have demonstrated an increase in acetate- and butyrate-producing bacteria such as Clostridium leptum, Bacteroides and Prevotella, which may be related to improved blood glucose and glucose intolerance. Five out of nine different probiotic strains tested in mice with high-fat diet and streptozoticin-induced type 2 diabetes increased short-chain fatty acids (SCFA)-producing gut bacteria such as Bacteroidales S24-7, Parabacteroides, Mucispirillum and Coprococcus. In addition, these strains have been directly associated with improving glucose metabolism and alleviating insulin resistance [77]. A correlation between plasma glucose and the Firmicutes (Bacillota)/Bacteroidetes (Bacteroidota) ratio has been proposed also by Yan et al. They showed that the administration of two strains of Lactobacillus acidophilus increased the relative abundance of the genera Blautia, Roseburia and Anaerotruncus, while Gram-negative bacteria of the genera Desulfovibrio, Alistipes and Bacteroides decreased [110]. Administration of heated-killed Streptococcus thermophilus improved some glycaemic parameters of diabetic rats, while increasing the levels of some beneficial bacteria of the genera Ruminococcaceae, Veillonella, Coprococcus and Barnesiella [84]. A reduction in blood glucose levels due to the enrichment of the genus Alistipes following the administration of engineered Lactobacillus plantarum-pMG36e-GLP-1 in diabetic monkeys has been suggested. Moreover, the probiotic reduced the phylum Bacteroidetes (Bacteroidota), particularly the bacterial species of Prevotella, whose association with type 2 diabetes has already been confirmed [200].
Zheng et al. [201] showed that probiotic supplementation of Lactobacillus rhamnosus LGG and Bifidobacterium animalis subsp. lactis Bb12 alleviated GDM in pregnant rats and showed an improvement in fasting blood glucose levels due to restoration of the gut microbiota. In fact, the abundance of Firmicutes (Bacillota) and Actinobacteria (Actinomycetota) increased. Furthermore, the probiotic mechanism related to GDM might be linked to the inhibition of carbohydrate metabolism and membrane transport pathways.

3.8.2. Effect of Probiotics on the Formation of SCFA

SCFA are non-digestive (human) metabolic nutrients derived mainly from the fermentation of resistant carbohydrates. In humans, SCFA account for 10% of daily energy expenditure and are predominantly of three types, acetic, propionic and butyric acids [202]. SCFA have been proposed as an important link between health and disease, particularly as energy carriers, modulators of cell proliferation and differentiation, and as immune modulators [203] and therefore as an important research component in analytical methods [204].
Yadav et al. [205] have shown in mouse models that the addition of the probiotic VSL#3 has a positive effect on obesity and diabetes. The probiotic reduced appetite, body weight and insulin resistance and increased glucose tolerance by modulating the composition of the gut microbiota and increased butyrate levels. Wang et al. [155] also hypothesised that the use of the probiotic Lactobacillus casei CCF419 influenced the regulation of the structure of the gut microbiota and thus the composition of SCFA in favour of increased acetate and butyrate. A very similar effect was observed in a study with high-fat and streptozotocin-induced diabetic mice. After administration of Lactobacillus plantarum HAC01, a restoration of the gut microbiota and an increase in butyric acid levels were observed [103]. Five different probiotic strains out of nine tested increased SCFA levels in the faeces of mice with high-fat diets and streptozoticin-induced type 2 diabetes, while showing a hypoglycaemic effect, reducing inflammation and decreasing insulin resistance. An increase in SCFA levels was associated with increased abundance in the SCFA-producing gut microbiota [77].
One of the potential mechanisms of SCFA in the prevention of type 2 diabetes involves intestinal gluconeogenesis, which is directly linked to the periportal nervous system via a specific signalling system. Thus, intestinal gluconeogenesis is inversely correlated with the risk factor for type 2 diabetes, as shown by several studies demonstrating a decrease in body weight, secretion of glucose in the liver and improved glycaemic control [24]. Furthermore, butyrate directly enhances the expression of genes involved in intestinal gluconeogenesis via free fatty acid receptors (FFAR). On the other hand, propionic acid (propionate) binds to FFAR3 in the portal vein nerves. Both signalling pathways are important for the glucose portal sensing process [206]. Toejing et al. [82] and Yan et al. [110] have looked at the importance of increased SCFA levels in the caecum of rats and mice for glucose metabolism after administration of Lactobacillus paracasei HII01 and Lactobacillus acidophilus, respectively.
GPR43 is one of the specific G protein-coupled receptors (GPRs) and it is known that its activation by acetate and propionate is more sensitive than by butyrate [207]. In a study in mice, Horiuchi et al. suggested that increased levels of SCFA (especially acetate) by administration of Bifidobacterium animalis subsp. lactis GCL2505 improved host glucose tolerance and energy expenditure and overcame body fat accumulation by activating GPR43 [208]. One of the several antidiabetic effects demonstrated in diabetic mice after administration of 14 compound probiotics was improved insulin secretion via glucose-triggered secretion of glucagon-like polypeptide (GLP)-1 and upregulation of GPR43/41 [100]. Lu et al. [209] suggested that a composite of three probiotics could act on tumour suppression in colorectal cancer cells by inhibiting tumourigenesis and glucose metabolism through GPR43 activation. However, the activation may have resulted in decreased expression of some glycolytic genes, which in turn resulted in decreased glucose metabolism.

3.8.3. Effect of Probiotics on the Secretion of Intestinal Incretins

Incretins are intestinal hormones secreted by enteroendocrine cells. The best-known representatives, GLP-1 and the glucose-dependent insulinotropic polypeptide, i.e., gastric inhibitory polypeptide (GIP), are involved in the physiological regulation of glucose homeostasis. When glucose increases, (e.g., due to food intake), the two hormones are released into the blood to increase the synthesis and release of insulin from pancreatic cells [210]. In addition, GLP-1 reduces the secretion of the hormone glucagon from the α-cells of the pancreas, which helps to control blood glucose levels. In addition, GLP-1 has an effect on decreased appetite, proliferation of pancreatic β-cells, gastrointestinal motility and gastric emptying. However, the endogenous enzyme dipeptidyl peptidase-4 (DPP-4) protease ensures rapid degradation of incretins in the body, which consequently affects their short-term activity in the blood. However, secretion of incretins can be significantly impaired in conditions such as obesity and type 2 diabetes [211].
The importance of GLP-1 in relation to blood glucose levels was demonstrated in a study on diabetic monkeys. Luo et al. showed the blood sugar-lowering effect after administration of a genetically modified strain Lactobacillus plantarum-pMG36e-GLP-1, which constantly expresses GLP-1 [200].
In a clinical trial with 21 subjects, it was shown that a 4-week intake of Lactobacillus reuteri SD586521 increased the secretion of GLP-1, which was associated with an increase in insulin and C-peptide concentrations. Therefore, the authors presented the probiotic Lactobacillus reuteri SD586521 as a new therapeutic approach to improve glucose-dependent insulin secretion [212].
Several studies in animals and in vitro models have demonstrated the effect of probiotics (of the species Lactobacillus) on increased incretin secretion (GLP-1) and consequently glucose control, as well as reducing signs of obesity and/or type 2 diabetes, i.e., postprandial glucose and insulin resistance [100,101,147,155,213,214].
In a study in rats, Yamano et al. [215] presented the potential mechanism of action of the probiotic Lactobacillus johnsonii La1. The probiotic appears to inhibit the activity of the adrenal sympathetic nervous system. They suggested that the lowering of blood glucose and glucagon concentrations was due to the probiotic altering the activity of the autonomic nervous system.
PDX-1, an insulin promoter (stimulating) factor 1, is involved in the glucose-dependent regulation of insulin gene transcription and is essential for the development of pancreatic exocrine and endocrine cells, including β-cells [216]. In an in vitro study, Duan et al. [217] demonstrated an insulinotropic effect of Escherichia coli Nissle 1917 on the secretion of GLP-1 and transcription activator PDX-1. The bacterial supernatant has been shown to stimulate intestinal epithelial cells to secrete insulin. Moreover, they also emphasised the importance of optimised conditions to achieve secretion of insulin levels, which is crucial for normal metabolism.
In an in vitro study, Belguesmia et al. [218] assumed that some strains of the genus Lactobacillus are able to degrade certain gut hormones through serine peptidase action. In this way, the probiotic can indirectly influence glucose metabolism and homeostasis.

3.8.4. Effect of Probiotics on Permeability and Integrity of the Intestinal Wall

Lim et al. [132] demonstrated in a mouse model that Lactobacillus sakei OK67 alleviated hyperglycaemia induced by a high-fat diet by reducing inflammation and increasing the expression of tight junction (TJ) proteins in the intestinal wall. Because probiotics affect the composition of the gut microflora, studies in rats [219,220] and mice [100,101,105,107,110] have shown that they improve the integrity of the intestinal epithelium and enhance the local immune response. In fact, the immunomodulatory effect of various probiotics led to a reduction in blood glucose levels. Moreover, in studies in rats [219,220] and a study in mice [105], probiotics inhibited intestinal toll-like receptor 4 (TLR 4) signalling [105,219,220] and activated toll-like receptor 2 (TLR 2) [105], which consequently had an effect on reducing some inflammatory processes, increasing insulin resistance and attenuating impaired glucose tolerance. In addition, decreased TLR 4 and increased TLR 2 mRNA levels were associated with decreased mRNA levels of the pro-inflammatory cytokines TNF-α and IL-6 [105].
An in vitro study with different Lactobacillus strains has shown that Lactobacillus gasseri ICVB396 contributes at most to the restoration and strengthening of the epithelial wall of the intestine in the Caco2 cell line [218].
TNF-α and IL-6 were investigated in an in vitro study of LPS-induced inflammation in Caco-2 cells as well as in the intestinal macrophage line RAW264.7 after addition of three different probiotic strains. A reduction in both pro-inflammatory parameters proved an improvement in intestinal restoration and insulin resistance [147].

3.8.5. Effect of Probiotics on Glucose Transport in Intestine

Rooj et al. [221] have demonstrated the interaction of Lactobacillus acidophilus and other species of the genus Lactobacillus with the intestinal epithelium in an in vitro study. After the short-term exposure of intestinal cells to a bacterial supernatant, reduced glucose accumulation was observed.
In CaCo-2 cells, Hsieh et al. [93] showed that Lactobacillus salivarus AP-32 and Lactobacillus reuteri GL-104 downregulated two major hexose transporters in the brush border gut membrane–sodium–glucose cotransporter 1 (SGLT1) and GLUT5. Consequently, both transporters automatically blocked sugar uptake in the gut. However, the expression of the basolateral glucose transporter GLUT2 was upregulated, leading to speculation that the upregulation of GLUT2 may have occurred in response to the reduction in SGLT1 and GLUT5 expression.
Another in vitro study on Caco-2 cells showed possible effects of three different strains on enhanced glucose uptake, possibly through upregulation of the PI3K/AKT pathway [147].
In our recent in vitro study [222] we tested the hypothesis of the effects of selected Lactobacillus probiotics on the regulation of the enterocytic transporters SGLT1 and GLUT2 in two different intestinal epithelial models. We demonstrated that the changes in expression of both transporters were strain- and cell-line-specific. Moreover, Lactobacillus plantarum PCS26 and Lactobacillus acidophilus downregulated GLUT2 expression, which was also observed by decreased transepithelial glucose transport rates, leading to reduction in glucose absorption.
Wang et al. showed that administration of eight probiotic strains decreased the expression of SGLT1 and GLUT2 in STZ-induced diabetic mice, indicating their effect on reducing digestion and glucose absorption in the gut [96].

4. Concluding Remarks and Future Perspectives

Our review shows that probiotics play an important role in health and diseases related to glucose metabolism. Numerous in vivo studies in humans and animals, as well as studies in vitro, have clearly shown that probiotics can exert their direct or indirect effects on glucose and its metabolism in almost all tissues and organs of the body. Moreover, their interaction with the physiological functions of various brain–hormone axes has proven to play an important role and act in several areas of the species’ body. Although most of the proposed probiotic effects have been shown to be beneficial and positive results have been obtained, there still seems to be a large amount of variation in results, especially between human studies, due to a variety of aspects on which one may or may not have specific effects.
A standardisation of methodological approaches in in vivo studies can definitely contribute to a better understanding of the potential probiotic mechanisms and the consequences of their effects. Therefore, the use of one or more strains (of the same), the administration doses (amount) and their duration, the number of patients/animals, their age, their diet and their possible physical activity (diversity within the study) should be considered more seriously before therapy. Furthermore, many methodological approaches still lack possible probiotic side effects and additional testing of other metabolic factors affecting glycaemic control. Given the above limitations, research into probiotic mechanisms and their potentially broader role in body functioning could be improved. Nevertheless, numerous effects of probiotics on glucose metabolism have been studied in various pathological metabolic conditions such as type 2 diabetes, metabolic syndrome, insulin resistance, glucose intolerance and hyperglycaemia. In this regard, since the initiation of any probiotic therapy, a disease state, its severity or potential risk factors associated with its outcome should be of enormous importance. In vivo animal models should consider whether induced or naturally occurring diseases were included in the study, as they may have different effects on probiotics and thus on their effects in animals. However, the animal studies appeared to be more homogeneous with regard to the effects of probiotics on glucose metabolism and homeostasis.
Nevertheless, there are some limitations in in vivo human studies that may always compromise the variety of probiotic effects, regardless of their potentially unique positive outcomes, especially in glucose metabolism and its homeostasis. Humans are to some extent genetically different and not alike due to environmental and geographical differences. They also have their own dietary habits that greatly affect their microbiota and probiotics as well. Therefore, more consideration should be given to the status of the microbiota, which is considered as unique as a fingerprint, before probiotic therapy. Last but not least, specific alterations of the microbiota have been associated with various pathological diseases affecting glucose metabolism and its homeostasis, and their modulation through probiotic administration could be important in studies on disease-targeted therapies. However, the number of human studies on this topic is still small, just as the studies on the exact effects (short- or long-term) of probiotics on the intestinal microbiota that influence glucose homeostasis are not fully presented and clarified. The fact that modification in intestinal microbiota after the administration of probiotics varies depending on the type of model (and its pathological/physiological conditions) and the experimental conditions is also something we have learned from the studies on animal models. Although more interactions between probiotics–intestinal microbiota–glucose metabolism and its homeostasis through the microbiota-related intestinal wall, specific microbiota-related SCFAs affecting hormone and SCFA secretion and glucose/lipid metabolism-related signalling routes are proposed, the exact mechanisms of how probiotics work are still being thoroughly investigated.
It has already been proven that probiotics have species- and strain-specific effects. In terms of effects on glucose metabolism and its homeostasis, studies with the genus Lactobacillus still predominate, followed by Bifidobacterium. However, studies on novel probiotic strains belonging to the genera of Akkermansia, Faecalibacterium, Prevotella have limited publications, but they have shown potential efficacy and should therefore be the subject of further future studies.
Moreover, further studies on the interactions between probiotics and the host are needed. Indeed, for proper interactions to occur, the involvement of commensals appears to be essential. Probiotics are microorganisms that serve to achieve a balanced co-existence and simultaneous synergistic effect with the host microbiota. The relationship between the two, as well as their effects on the host immune system, are therefore unique and should be studied as such when it comes to new research on the probiotics mechanisms of action.
Since energy metabolism is related to gluconeogenesis and the production of ketone bodies during starvation or diets with very low availability of digestible carbohydrates [223], more importance of analysing ketone bodies in the context of hypo- and more recently hyperglycaemic states in in vivo studies should be considered. Furthermore, the effect of probiotics on ketone body levels and energy metabolism reduction as well as their positive correlation, has been demonstrated in animal models [224].
Although the use of probiotics and prebiotics as modulators of the intestinal microbiota on the host still has the most successful short- and long-term effects, especially in glucose-related diseases, faecal microbiota transplantation has raised many promising expectations in this field [225,226]. Short-term improvement in some parameters of glucose and lipid metabolism has been reported [227,228,229], but the long-term effects in terms of their side effects still need to be carefully studied, as it seems that more than one treatment should be considered to achieve adequate results.

Author Contributions

Conceptualisation, M.P. and T.L.; methodology, M.P.; investigation, M.P.; resources, M.P.; data curation, M.P.; writing—original draft preparation, M.P.; writing—review and editing, T.L.; visualisation, T.L.; supervision, T.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Slovenian Research Agency (Javna Agencija za Raziskovalno Dejavnost RS), grant number P1-0164 (C).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

We would like to thank Martin Kozmos for graphical assistance.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Harris, R.A. Carbohydrate metabolism I: Major metabolic pathways and their control. In Textbook of Biochemistry with Clinical Correlations, 4th ed.; Devlin, T.M., Ed.; Wiley, Inc.: New York, NY, USA, 1997; pp. 267–335. [Google Scholar]
  2. Simon, K.; Wittmann, I. Can blood glucose value really be referred to as a metabolic parameter? Rev. Endocr. Metab. Disord. 2019, 20, 151–160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Lee, S.H.; Park, S.Y.; Choi, C.S. Insulin Resistance: From Mechanisms to Therapeutic Strategies. Diabetes Metab. J. 2022, 46, 15–37. [Google Scholar] [CrossRef] [PubMed]
  4. Rohm, T.V.; Meier, D.T.; Olefsky, J.M.; Donath, M.Y. Inflammation in obesity, diabetes, and related disorders. Immunity 2022, 55, 31–55. [Google Scholar] [CrossRef] [PubMed]
  5. Lima, J.E.B.F.; Moreira, N.C.S.; Sakamoto-Hojo, E.T. Mechanisms underlying the pathophysiology of type 2 diabetes: From risk factors to oxidative stress, metabolic dysfunction, and hyperglycemia. Mutat. Res. Genet. Toxicol. Environ. Mutagen. 2022, 874–875, 503437. [Google Scholar] [CrossRef] [PubMed]
  6. Ceriello, A. Postprandial Hyperglycemia and Diabetes Complications: Is It Time to Treat? Diabetes 2005, 54, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Mouri, M.; Badireddy, M. Hyperglycemia; StatPearls Publishing: Treasure Island, FL, USA, 2019. [Google Scholar]
  8. Giri, B.; Dey, S.; Das, T.; Sarkar, M.; Banerjee, J.; Dash, S.K. Chronic hyperglycemia mediated physiological alteration and metabolic distortion leads to organ dysfunction, infection, cancer progression and other pathophysiological consequences: An update on glucose toxicity. Biomed. Pharmacother. 2018, 107, 306–328. [Google Scholar] [CrossRef]
  9. Nishida, C.; Uauy, R.; Kumanyika, S.; Shetty, P. The joint WHO/FAO expert consultation on diet, nutrition and the prevention of chronic diseases: Process, product and policy implications. Public Health Nutr. 2004, 7, 245–250. [Google Scholar] [CrossRef] [Green Version]
  10. World Health Organization. Diet, Nutrition, and the Prevention of Chronic Diseases: Report of a WHO Study Group; World Health Organization: Geneva, Switzerland, 1990; ISBN 9241207973. [Google Scholar]
  11. Tuomilehto, J.; Lindström, J.; Eriksson, J.G.; Valle, T.T.; Hämäläinen, H.; Ilanne-Parikka, P.; Keinänen-Kiukaanniemi, S.; Laakso, M.; Louheranta, A.; Rastas, M.; et al. Prevention of Type 2 Diabetes Mellitus by Changes in Lifestyle among Subjects with Impaired Glucose Tolerance. N. Engl. J. Med. 2001, 344, 1343–1350. [Google Scholar] [CrossRef]
  12. Andrade, E.F.; de Oliveira Silva, V.; Orlando, D.R.; Pereira, L.J. Mechanisms Involved in Glycemic Control Promoted by Exercise in Diabetics. Curr. Diabetes Rev. 2019, 15, 105–110. [Google Scholar] [CrossRef]
  13. Gérard, C.; Vidal, H. Impact of Gut Microbiota on Host Glycemic Control. Front. Endocrinol. 2019, 10, 29. [Google Scholar] [CrossRef]
  14. Sivamaruthi, B.S.; Kesika, P.; Suganthy, N.; Chaiyasut, C. A Review on Role of Microbiome in Obesity and Antiobesity Properties of Probiotic Supplements. Biomed Res. Int. 2019, 2019, 3291367. [Google Scholar] [CrossRef]
  15. Tang, C.; Kong, L.; Shan, M.; Lu, Z.; Lu, Y. Protective and ameliorating effects of probiotics against diet-induced obesity: A review. Food Res. Int. 2021, 147, 110490. [Google Scholar] [CrossRef]
  16. Torres, S.; Fabersani, E.; Marquez, A.; Gauffin-Cano, P. Adipose tissue inflammation and metabolic syndrome. The proactive role of probiotics. Eur. J. Nutr. 2019, 58, 27–43. [Google Scholar] [CrossRef]
  17. He, M.; Shi, B. Gut microbiota as a potential target of metabolic syndrome: The role of probiotics and prebiotics. Cell Biosci. 2017, 7, 54. [Google Scholar] [CrossRef] [Green Version]
  18. Wang, G.; Liu, J.; Xia, Y.; Ai, L. Probiotics-based interventions for diabetes mellitus: A review. Food Biosci. 2021, 43, 101172. [Google Scholar] [CrossRef]
  19. Sun, Z.; Sun, X.; Li, J.; Li, Z.; Hu, Q.; Li, L.; Hao, X.; Song, M.; Li, C. Using probiotics for type 2 diabetes mellitus intervention: Advances, questions, and potential. Crit. Rev. Food Sci. Nutr. 2019, 60, 670–683. [Google Scholar] [CrossRef]
  20. Zhai, L.; Wu, J.; Lam, Y.Y.; Kwan, H.Y.; Bian, Z.X.; Wong, H.L.X. Gut-Microbial Metabolites, Probiotics and Their Roles in Type 2 Diabetes. Int. J. Mol. Sci. 2021, 22, 12846. [Google Scholar] [CrossRef]
  21. Iatcu, C.O.; Steen, A.; Covasa, M. Gut Microbiota and Complications of Type-2 Diabetes. Nutrients 2021, 14, 166. [Google Scholar] [CrossRef] [PubMed]
  22. Cani, P.D.; Amar, J.; Iglesias, M.A.; Poggi, M.; Knauf, C.; Bastelica, D.; Neyrinck, A.M.; Fava, F.; Tuohy, K.M.; Chabo, C.; et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007, 56, 1761–1772. [Google Scholar] [CrossRef] [Green Version]
  23. Salari, A.; Mahdavi-Roshan, M.; Kheirkhah, J.; Ghorbani, Z. Probiotics supplementation and cardiometabolic risk factors: A new insight into recent advances, potential mechanisms, and clinical implications. PharmaNutrition 2021, 16, 100261. [Google Scholar] [CrossRef]
  24. Kim, Y.A.; Keogh, J.B.; Clifton, P.M. Probiotics, prebiotics, synbiotics and insulin sensitivity. Nutr. Res. Rev. 2018, 31, 35–51. [Google Scholar] [CrossRef] [PubMed]
  25. Miglioranza Scavuzzi, B.; Miglioranza, L.H.d.S.; Henrique, F.C.; Pitelli Paroschi, T.; Lozovoy, M.A.B.; Simão, A.N.C.; Dichi, I. The role of probiotics on each component of the metabolic syndrome and other cardiovascular risks. Expert Opin. Ther. Targets 2015, 19, 1127–1138. [Google Scholar] [CrossRef] [PubMed]
  26. Razmpoosh, E.; Javadi, M.; Ejtahed, H.-S.; Mirmiran, P. Probiotics as beneficial agents in the management of diabetes mellitus: A systematic review. Diabetes. Metab. Res. Rev. 2016, 32, 143–168. [Google Scholar] [CrossRef] [PubMed]
  27. Bezirtzoglou, E.; Stavropoulou, E.; Kantartzi, K.; Tsigalou, C.; Voidarou, C.; Mitropoulou, G.; Prapa, I.; Santarmaki, V.; Kompoura, V.; Yanni, A.E.; et al. Maintaining Digestive Health in Diabetes: The Role of the Gut Microbiome and the Challenge of Functional Foods. Microorganisms 2021, 9, 516. [Google Scholar] [CrossRef]
  28. Papadopoulos, P.D.; Tsigalou, C.; Valsamaki, P.N.; Konstantinidis, T.G.; Voidarou, C.; Bezirtzoglou, E. The Emerging Role of the Gut Microbiome in Cardiovascular Disease: Current Knowledge and Perspectives. Biomedicines 2022, 10, 948. [Google Scholar] [CrossRef]
  29. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef] [Green Version]
  30. FAO/WHO Guidelines for the Evaluation of Probiotics in Food. Report of a Joint FAO/WHO Working Group on Drafting Guidelines for the Evaluation of Probiotics in Food. Available online: http://www.who.int/foodsafety/fs_management/en/probiotic_guidelines.pdf?ua=1 (accessed on 23 May 2019).
  31. EFSA Opinion of the Scientific Committee on a request from EFSA related to a generic approach to the safety assessment by EFSA of microorganisms used in food/feed and the production of food/feed additives. EFSA J. 2005, 226, 1–12.
  32. Zheng, J.; Wittouck, S.; Salvetti, E.; Franz, C.M.A.P.; Harris, H.M.B.; Mattarelli, P.; O’Toole, P.W.; Pot, B.; Vandamme, P.; Walter, J.; et al. A taxonomic note on the genus Lactobacillus: Description of 23 novel genera, emended description of the genus Lactobacillus Beijerinck 1901, and union of Lactobacillaceae and Leuconostocaceae. Int. J. Syst. Evol. Microbiol. 2020, 70, 2782–2858. [Google Scholar] [CrossRef]
  33. Voidarou, C.; Antoniadou, M.; Rozos, G.; Tzora, A.; Skoufos, I.; Varzakas, T.; Lagiou, A.; Bezirtzoglou, E. Fermentative Foods: Microbiology, Biochemistry, Potential Human Health Benefits and Public Health Issues. Foods 2020, 10, 69. [Google Scholar] [CrossRef]
  34. Hampe, C.S.; Roth, C.L. Probiotic strains and mechanistic insights for the treatment of type 2 diabetes. Endocrine 2017, 58, 207–227. [Google Scholar] [CrossRef]
  35. George Kerry, R.; Patra, J.K.; Gouda, S.; Park, Y.; Shin, H.-S.; Das, G. Benefaction of probiotics for human health: A review. J. Food Drug Anal. 2018, 26, 927–939. [Google Scholar] [CrossRef] [Green Version]
  36. De Almada, C.N.; Almada, C.N.; Martinez, R.C.R.; Sant’Ana, A.S. Paraprobiotics: Evidences on their ability to modify biological responses, inactivation methods and perspectives on their application in foods. Trends Food Sci. Technol. 2016, 58, 96–114. [Google Scholar] [CrossRef]
  37. Turpin, W.; Humblot, C.; Thomas, M.; Guyot, J.-P. Lactobacilli as multifaceted probiotics with poorly disclosed molecular mechanisms. Int. J. Food Microbiol. 2010, 143, 87–102. [Google Scholar] [CrossRef]
  38. Reid, G. Probiotics: Definition, scope and mechanisms of action. Best Pract. Res. Clin. Gastroenterol. 2016, 30, 17–25. [Google Scholar] [CrossRef]
  39. Peterson, C.T.; Sharma, V.; Elmén, L.; Peterson, S.N. Immune homeostasis, dysbiosis and therapeutic modulation of the gut microbiota. Clin. Exp. Immunol. 2015, 179, 363–377. [Google Scholar] [CrossRef] [Green Version]
  40. Plaza-Diaz, J.; Ruiz-Ojeda, F.J.; Gil-Campos, M.; Gil, A. Mechanisms of Action of Probiotics. Adv. Nutr. 2019, 10, S49–S66. [Google Scholar] [CrossRef] [Green Version]
  41. Suez, J.; Zmora, N.; Segal, E.; Elinav, E. The pros, cons, and many unknowns of probiotics. Nat. Med. 2019, 25, 716–729. [Google Scholar] [CrossRef]
  42. Barros, C.P.; Grom, L.C.; Guimarães, J.T.; Balthazar, C.F.; Rocha, R.S.; Silva, R.; Almada, C.N.; Pimentel, T.C.; Venâncio, E.L.; Collopy Junior, I.; et al. Paraprobiotic obtained by ohmic heating added in whey-grape juice drink is effective to control postprandial glycemia in healthy adults. Food Res. Int. 2021, 140, 109905. [Google Scholar] [CrossRef]
  43. Ejtahed, H.S.; Mohtadi-Nia, J.; Homayouni-Rad, A.; Niafar, M.; Asghari-Jafarabadi, M.; Mofid, V. Probiotic yogurt improves antioxidant status in type 2 diabetic patients. Nutrition 2012, 28, 539–543. [Google Scholar] [CrossRef]
  44. 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]
  45. Schellekens, H.; Torres-Fuentes, C.; van de Wouw, M.; Long-Smith, C.M.; Mitchell, A.; Strain, C.; Berding, K.; Bastiaanssen, T.F.S.; Rea, K.; Golubeva, A.V.; et al. Bifidobacterium longum counters the effects of obesity: Partial successful translation from rodent to human. EBioMedicine 2021, 63, 103176. [Google Scholar] [CrossRef]
  46. Toshimitsu, T.; Gotou, A.; Furuichi, K.; Hachimura, S.; Asami, Y. Effects of 12-wk Lactobacillus plantarum OLL2712 treatment on glucose metabolism and chronic inflammation in prediabetic individuals: A single-arm pilot study. Nutrition 2019, 58, 175–180. [Google Scholar] [CrossRef]
  47. Sabico, S.; Al-Mashharawi, A.; Al-Daghri, N.M.; Wani, K.; Amer, O.E.; Hussain, D.S.; Ahmed Ansari, M.G.; Masoud, M.S.; Alokail, M.S.; McTernan, P.G. Effects of a 6-month multi-strain probiotics supplementation in endotoxemic, inflammatory and cardiometabolic status of T2DM patients: A randomized, double-blind, placebo-controlled trial. Clin. Nutr. 2019, 38, 1561–1569. [Google Scholar] [CrossRef] [Green Version]
  48. Khalili, L.; Alipour, B.; Jafar-Abadi, M.A.; Faraji, I.; Hassanalilou, T.; Abbasi, M.M.; Vaghef-Mehrabany, E.; Sani, M.A. The Effects of Lactobacillus casei on Glycemic Response, Serum Sirtuin1 and Fetuin-A Levels in Patients with Type 2 Diabetes Mellitus: A Randomized Controlled Trial. Iran. Biomed. J. 2019, 23, 68–77. [Google Scholar] [CrossRef] [Green Version]
  49. Khalili, L.; Alipour, B.; Asghari Jafarabadi, M.; Hassanalilou, T.; Mesgari Abbasi, M.; Faraji, I. Probiotic assisted weight management as a main factor for glycemic control in patients with type 2 diabetes: A randomized controlled trial. Diabetol. Metab. Syndr. 2019, 11, 5. [Google Scholar] [CrossRef]
  50. Tonucci, L.B.; Olbrich dos Santos, K.M.; Licursi de Oliveira, L.; Rocha Ribeiro, S.M.; Duarte Martino, H.S. Clinical application of probiotics in type 2 diabetes mellitus: A randomized, double-blind, placebo-controlled study. Clin. Nutr. 2017, 36, 85–92. [Google Scholar] [CrossRef]
  51. Hsieh, M.C.; Tsai, W.H.; Jheng, Y.P.; Su, S.L.; Wang, S.Y.; Lin, C.C.; Chen, Y.H.; Chang, W.W. The beneficial effects of Lactobacillus reuteri ADR-1 or ADR-3 consumption on type 2 diabetes mellitus: A randomized, double-blinded, placebo-controlled trial. Sci. Rep. 2018, 8, 16791. [Google Scholar] [CrossRef]
  52. 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. Clin. Res. Rev. 2018, 12, 617–624. [Google Scholar] [CrossRef] [PubMed]
  53. 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]
  54. 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]
  55. Andreasen, A.S.; Larsen, N.; Pedersen-Skovsgaard, T.; Berg, R.M.G.; Møller, K.; Svendsen, K.D.; Jakobsen, M.; Pedersen, B.K. Effects of Lactobacillus acidophilus NCFM on insulin sensitivity and the systemic inflammatory response in human subjects. Br. J. Nutr. 2010, 104, 1831–1838. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Hulston, C.J.; Churnside, A.A.; Venables, M.C. Probiotic supplementation prevents high-fat, overfeeding-induced insulin resistance in human subjects. Br. J. Nutr. 2015, 113, 596–602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Chapman, C.M.C.; Gibson, G.R.; Rowland, I. Health benefits of probiotics: Are mixtures more effective than single strains? Eur. J. Nutr. 2011, 50, 1–17. [Google Scholar] [CrossRef] [PubMed]
  58. Kurl, S.; Zaccardi, F.; Onaemo, V.N.; Jae, S.Y.; Kauhanen, J.; Ronkainen, K.; Laukkanen, J.A. Association between HOMA-IR, fasting insulin and fasting glucose with coronary heart disease mortality in nondiabetic men: A 20-year observational study. Acta Diabetol. 2015, 52, 183–186. [Google Scholar] [CrossRef]
  59. Holmes, D. Physiologic role of IL-1β in glucose homeostasis. Nat. Rev. Endocrinol. 2017, 13, 128. [Google Scholar] [CrossRef]
  60. Ibfelt, T.; Fischer, C.P.; Plomgaard, P.; Van Hall, G.; Pedersen, B.K. The acute effects of low-dose TNF- on glucose metabolism and β -cell function in humans. Mediators Inflamm. 2014, 2014, 295478. [Google Scholar] [CrossRef]
  61. Gonzalez, Y.; Herrera, M.T.; Soldevila, G.; Garcia-Garcia, L.; Fabián, G.; Pérez-Armendariz, E.M.; Bobadilla, K.; Guzmán-Beltrán, S.; Sada, E.; Torres, M. High glucose concentrations induce TNF-α production through the down-regulation of CD33 in primary human monocytes. BMC Immunol. 2012, 13, 19. [Google Scholar] [CrossRef] [Green Version]
  62. Hsieh, P.-S.; An, Y.; Tsai, Y.-C.; Chen, Y.-C.; Chuang, C.-J.; Zeng, C.-T.; Wang, C.-T.; An-Erl King, V. Potential of probiotic strains to modulate the inflammatory responses of epithelial and immune cells in vitro. New Microbiol. 2013, 36, 167–179. [Google Scholar]
  63. Asemi, Z.; Zare, Z.; Shakeri, H.; Sabihi, S.; Esmaillzadeh, A. Effect of Multispecies Probiotic Supplements on Metabolic Profiles, hs-CRP, and Oxidative Stress in Patients with Type 2 Diabetes. Ann. Nutr. Metab. 2013, 63, 1–9. [Google Scholar] [CrossRef]
  64. Ismail, N.A.; Ragab, S.; Abd El Dayem, S.M.; Abd ElBaky, A.; Salah, N.; Hamed, M.; Assal, H.; Koura, H. Fetuin-A levels in obesity: Differences in relation to metabolic syndrome and correlation with clinical and laboratory variables. Arch. Med. Sci. 2012, 8, 826–833. [Google Scholar] [CrossRef]
  65. Srinivas, P.R.; Wagner, A.S.; Reddy, L.V.; Deutsch, D.D.; Leon, M.A.; Goustin, A.S.; Grunberger, G. Serum alpha 2-HS-glycoprotein is an inhibitor of the human insulin receptor at the tyrosine kinase level. Mol. Endocrinol. 1993, 7, 1445–1455. [Google Scholar] [CrossRef] [Green Version]
  66. Turkmen, K.; Karagoz, A.; Kucuk, A. Sirtuins as novel players in the pathogenesis of diabetes mellitus. World J. Diabetes 2014, 5, 894. [Google Scholar] [CrossRef]
  67. Sun, J.; Buys, N.J. Glucose- and glycaemic factor-lowering effects of probiotics on diabetes: A meta-analysis of randomised placebo-controlled trials. Br. J. Nutr. 2016, 115, 1167–1177. [Google Scholar] [CrossRef]
  68. Zhang, Q.; Wu, Y.; Fei, X. Effect of probiotics on glucose metabolism in patients with type 2 diabetes mellitus: A meta-analysis of randomized controlled trials. Medicina 2016, 52, 28–34. [Google Scholar] [CrossRef]
  69. Samah, S.; Ramasamy, K.; Lim, S.M.; Neoh, C.F. Probiotics for the management of type 2 diabetes mellitus: A systematic review and meta-analysis. Diabetes Res. Clin. Pract. 2016, 118, 172–182. [Google Scholar] [CrossRef]
  70. Nikbakht, E.; Khalesi, S.; Singh, I.; Williams, L.T.; West, N.P.; Colson, N. Effect of probiotics and synbiotics on blood glucose: A systematic review and meta-analysis of controlled trials. Eur. J. Nutr. 2018, 57, 95–106. [Google Scholar] [CrossRef]
  71. Ruan, Y.; Sun, J.; He, J.; Chen, F.; Chen, R.; Chen, H. Effect of Probiotics on Glycemic Control: A Systematic Review and Meta-Analysis of Randomized, Controlled Trials. PLoS ONE 2015, 10, e0132121. [Google Scholar] [CrossRef] [Green Version]
  72. Li, C.; Li, X.; Han, H.; Cui, H.; Peng, M.; Wang, G.; Wang, Z. Effect of probiotics on metabolic profiles in type 2 diabetes mellitus. Medicine 2016, 95, e4088. [Google Scholar] [CrossRef]
  73. He, J.; Zhang, F.; Han, Y. Effect of probiotics on lipid profiles and blood pressure in patients with type 2 diabetes: A meta-analysis of RCTs. Medicine 2017, 96, e9166. [Google Scholar] [CrossRef]
  74. Kasińska, M.A.; Drzewoski, J. Effectiveness of probiotics in type 2 diabetes: A meta-analysis. Pol. Arch. Med. Wewn. 2015, 125, 803–813. [Google Scholar] [CrossRef] [Green Version]
  75. Ardeshirlarijani, E.; Tabatabaei-Malazy, O.; Mohseni, S.; Qorbani, M.; Larijani, B.; Baradar Jalili, R. Effect of probiotics supplementation on glucose and oxidative stress in type 2 diabetes mellitus: A meta-analysis of randomized trials. DARU J. Pharm. Sci. 2019, 27, 827–837. [Google Scholar] [CrossRef]
  76. Tao, Y.-W.; Gu, Y.-L.; Mao, X.-Q.; Zhang, L.; Pei, Y.-F. Effects of probiotics on type II diabetes mellitus: A meta-analysis. J. Transl. Med. 2020, 18, 30. [Google Scholar] [CrossRef] [Green Version]
  77. Wang, G.; Si, Q.; Yang, S.; Jiao, T.; Zhu, H.; Tian, P.; Wang, L.; Li, X.; Gong, L.; Zhao, J.; et al. Lactic acid bacteria reduce diabetes symptoms in mice by alleviating gut microbiota dysbiosis and inflammation in different manners. Food Funct. 2020, 11, 5898–5914. [Google Scholar] [CrossRef]
  78. Li, X.; Wang, N.; Yin, B.; Fang, D.; Jiang, T.; Fang, S.; Zhao, J.; Zhang, H.; Wang, G.; Chen, W. Effects of Lactobacillus plantarum CCFM0236 on hyperglycaemia and insulin resistance in high-fat and streptozotocin-induced type 2 diabetic mice. J. Appl. Microbiol. 2016, 121, 1727–1736. [Google Scholar] [CrossRef]
  79. Sharma, P.; Bhardwaj, P.; Singh, R. Administration of Lactobacillus casei and Bifidobacterium bifidum Ameliorated Hyperglycemia, Dyslipidemia, and Oxidative Stress in Diabetic Rats. Int. J. Prev. Med. 2016, 7, 102. [Google Scholar] [CrossRef]
  80. Bejar, W.; Hamden, K.; Ben Salah, R.; Chouayekh, H. Lactobacillus plantarum TN627 significantly reduces complications of alloxan-induced diabetes in rats. Anaerobe 2013, 24, 4–11. [Google Scholar] [CrossRef]
  81. Zhang, J.; Wang, S.; Zeng, Z.; Qin, Y.; Shen, Q.; Li, P. Anti-diabetic effects of Bifidobacterium animalis 01 through improving hepatic insulin sensitivity in type 2 diabetic rat model. J. Funct. Foods 2020, 67, 103843. [Google Scholar] [CrossRef]
  82. Toejing, P.; Khat-Udomkiri, N.; Intakhad, J.; Sirilun, S.; Chaiyasut, C.; Lailerd, N. Putative Mechanisms Responsible for the Antihyperglycemic Action of Lactobacillus paracasei HII01 in Experimental Type 2 Diabetic Rats. Nutrients 2020, 12, 3015. [Google Scholar] [CrossRef]
  83. Farida, E.; Nuraida, L.; Giriwono, P.E.; Jenie, B.S.L. Lactobacillus rhamnosus Reduces Blood Glucose Level through Downregulation of Gluconeogenesis Gene Expression in Streptozotocin-Induced Diabetic Rats. Int. J. Food Sci. 2020, 2020, 6108575. [Google Scholar] [CrossRef] [Green Version]
  84. Gao, X.; Wang, F.; Zhao, P.; Zhang, R.; Zeng, Q. Effect of heat-killed Streptococcus thermophilus on type 2 diabetes rats. PeerJ 2019, 7, e7117. [Google Scholar] [CrossRef] [Green Version]
  85. Micioni Di Bonaventura, M.V.; Coman, M.M.; Tomassoni, D.; Di Bonaventura, E.M.; Botticelli, L.; Gabrielli, M.G.; Rossolini, G.M.; Di Pilato, V.; Cecchini, C.; Amedei, A.; et al. Supplementation with Lactiplantibacillus plantarum IMC 510 Modifies Microbiota Composition and Prevents Body Weight Gain Induced by Cafeteria Diet in Rats. Int. J. Mol. Sci. 2021, 22, 11171. [Google Scholar] [CrossRef] [PubMed]
  86. Kumar, N.; Tomar, S.K.; Thakur, K.; Singh, A.K. The ameliorative effects of probiotic Lactobacillus fermentum strain RS-2 on alloxan induced diabetic rats. J. Funct. Foods 2017, 28, 275–284. [Google Scholar] [CrossRef]
  87. Xiaojie, L.; Yue, J.; Xiaofei, Z.; Naisheng, Z.; Jiandong, T.; Yongguo, C. Bacillus licheniformis Zhengchangsheng® Inhibits Obesity by Regulating the AMP-Activated Protein Kinase Signaling Pathway. Probiotics Antimicrob. Proteins 2021, 13, 1658–1667. [Google Scholar] [CrossRef]
  88. Choi, S.-I.; You, S.; Kim, S.; Won, G.; Kang, C.-H.; Kim, G.-H. Weissella cibaria MG5285 and Lactobacillus reuteri MG5149 attenuated fat accumulation in adipose and hepatic steatosis in high-fat diet-induced C57BL/6J obese mice. Food Nutr. Res. 2021, 65, 8087. [Google Scholar] [CrossRef] [PubMed]
  89. Hsieh, P.S.; Ho, H.-H.; Tsao, S.P.; Hsieh, S.-H.; Lin, W.-Y.; Chen, J.-F.; Kuo, Y.-W.; Tsai, S.-Y.; Huang, H.-Y. Multi-strain probiotic supplement attenuates streptozotocin-induced type-2 diabetes by reducing inflammation and β-cell death in rats. PLoS ONE 2021, 16, e0251646. [Google Scholar] [CrossRef] [PubMed]
  90. Okyere, S.K.; Xie, L.; Wen, J.; Ran, Y.; Ren, Z.; Deng, J.; Hu, Y. Bacillus toyonensis SAU-19 Ameliorates Hepatic Insulin Resistance in High-Fat Diet/Streptozocin-Induced Diabetic Mice. Nutritions 2021, 13, 4512. [Google Scholar] [CrossRef]
  91. Zhang, Y.; Wu, T.; Li, W.; Zhao, Y.; Long, H.; Liu, R.; Sui, W.; Zhang, M. Lactobacillus casei LC89 exerts antidiabetic effects through regulating hepatic glucagon response and gut microbiota in type 2 diabetic mice. Food Funct. 2021, 12, 8288–8299. [Google Scholar] [CrossRef]
  92. Holowacz, S.; Guigné, C.; Chêne, G.; Mouysset, S.; Guilbot, A.; Seyrig, C.; Dubourdeau, M. A multispecies Lactobacillus- and Bifidobacterium-containing probiotic mixture attenuates body weight gain and insulin resistance after a short-term challenge with a high-fat diet in C57/BL6J mice. PharmaNutrition 2015, 3, 101–107. [Google Scholar] [CrossRef] [Green Version]
  93. Hsieh, P.-S.; Ho, H.-H.; Hsieh, S.-H.; Kuo, Y.-W.; Tseng, H.-Y.; Kao, H.-F.; Wang, J.-Y. Lactobacillus salivarius AP-32 and Lactobacillus reuteri GL-104 decrease glycemic levels and attenuate diabetes-mediated liver and kidney injury in db/db mice. BMJ Open Diabetes Res. Care 2020, 8, e001028. [Google Scholar] [CrossRef] [Green Version]
  94. Zhong, H.; Zhang, Y.; Zhao, M.; Zhang, J.; Zhang, H.; Xi, Y.; Cai, H.; Feng, F. Screening of novel potential antidiabetic Lactobacillus plantarum strains based on in vitro and in vivo investigations. LWT 2021, 139, 110526. [Google Scholar] [CrossRef]
  95. Ben Othman, M.; Sakamoto, K. Effect of inactivated Bifidobacterium longum intake on obese diabetes model mice (TSOD). Food Res. Int. 2020, 129, 108792. [Google Scholar] [CrossRef]
  96. Wang, L.; Shang, Q.; Guo, W.; Wu, X.; Wu, L.; Wu, L.; Chen, T. Evaluation of the hypoglycemic effect of probiotics via directly consuming glucose in intestines of STZ-induced diabetic mice and glucose water-induced diabetic mice. J. Funct. Foods 2020, 64, 103614. [Google Scholar] [CrossRef]
  97. Andersson, U.; Bränning, C.; Ahrné, S.; Molin, G.; Alenfall, J.; Önning, G.; Nyman, M.; Holm, C. Probiotics lower plasma glucose in the high-fat fed C57BL/6J mouse. Benef. Microbes 2010, 1, 189–196. [Google Scholar] [CrossRef]
  98. Sakai, T.; Taki, T.; Nakamoto, A.; Shuto, E.; Tsutsumi, R.; Toshimitsu, T.; Makino, S.; Ikegami, S. Lactobacillus plantarum OLL2712 regulates glucose metabolism in C57BL/6 mice fed a high-fat diet. J. Nutr. Sci. Vitaminol. 2013, 59, 144–147. [Google Scholar] [CrossRef] [Green Version]
  99. Naito, E.; Yoshida, Y.; Makino, K.; Kounoshi, Y.; Kunihiro, S.; Takahashi, R.; Matsuzaki, T.; Miyazaki, K.; Ishikawa, F. Beneficial effect of oral administration of Lactobacillus casei strain Shirota on insulin resistance in diet-induced obesity mice. J. Appl. Microbiol. 2011, 110, 650–657. [Google Scholar] [CrossRef]
  100. Wang, Y.; Dilidaxi, D.; Wu, Y.; Sailike, J.; Sun, X.; Nabi, X. Composite probiotics alleviate type 2 diabetes by regulating intestinal microbiota and inducing GLP-1 secretion in db/db mice. Biomed. Pharmacother. 2020, 125, 109914. [Google Scholar] [CrossRef]
  101. Wang, Y.; Wu, Y.; Sailike, J.; Sun, X.; Abuduwaili, N.; Tuoliuhan, H.; Yusufu, M.; Nabi, X. Fourteen composite probiotics alleviate type 2 diabetes through modulating gut microbiota and modifying M1/M2 phenotype macrophage in db/db mice. Pharmacol. Res. 2020, 161, 105150. [Google Scholar] [CrossRef]
  102. Machado, A.S.; Oliveira, J.R.; Lelis, D.d.F.; de Paula, A.M.B.; Guimarães, A.L.S.; Andrade, J.M.O.; Brandi, I.V.; Santos, S.H.S. Oral Probiotic Bifidobacterium Longum Supplementation Improves Metabolic Parameters and Alters the Expression of the Renin-Angiotensin System in Obese Mice Liver. Biol. Res. Nurs. 2020, 23, 100–108. [Google Scholar] [CrossRef]
  103. Lee, Y.-S.; Lee, D.; Park, G.-S.; Ko, S.-H.; Park, J.; Lee, Y.-K.; Kang, J. Lactobacillus plantarum HAC01 ameliorates type 2 diabetes in high-fat diet and streptozotocin-induced diabetic mice in association with modulating the gut microbiota. Food Funct. 2021, 12, 6363–6373. [Google Scholar] [CrossRef]
  104. Zeng, Z.; Yuan, Q.; Yu, R.; Zhang, J.; Ma, H.; Chen, S. Ameliorative Effects of Probiotic Lactobacillus paracasei NL41 on Insulin Sensitivity, Oxidative Stress, and Beta-Cell Function in a Type 2 Diabetes Mellitus Rat Model. Mol. Nutr. Food Res. 2019, 63, 1900457. [Google Scholar] [CrossRef]
  105. Ashrafian, F.; Shahriary, A.; Behrouzi, A.; Moradi, H.R.; Keshavarz Azizi Raftar, S.; Lari, A.; Hadifar, S.; Yaghoubfar, R.; Ahmadi Badi, S.; Khatami, S.; et al. Akkermansia muciniphila-Derived Extracellular Vesicles as a Mucosal Delivery Vector for Amelioration of Obesity in Mice. Front. Microbiol. 2019, 10, 2155. [Google Scholar] [CrossRef] [PubMed]
  106. Wang, X.; Ba, T.; Cheng, Y.; Zhang, P.; Chang, X. Probiotics alleviate adipose inflammation in high-fat diet-induced obesity by restoring adipose invariant natural killer T cells. Nutrition 2021, 89, 111285. [Google Scholar] [CrossRef] [PubMed]
  107. Tarrah, A.; dos Santos Cruz, B.C.; Sousa Dias, R.; da Silva Duarte, V.; Pakroo, S.; Licursi de Oliveira, L.; Gouveia Peluzio, M.C.; Corich, V.; Giacomini, A.; Oliveira de Paula, S. Lactobacillus paracasei DTA81, a cholesterol-lowering strain having immunomodulatory activity, reveals gut microbiota regulation capability in BALB/c mice receiving high-fat diet. J. Appl. Microbiol. 2021, 131, 1942–1957. [Google Scholar] [CrossRef] [PubMed]
  108. Sun, Q.; Zhang, Y.; Li, Z.; Yan, H.; Li, J.; Wan, X. Mechanism analysis of improved glucose homeostasis and cholesterol metabolism in high-fat-induced obese mice treated with La-SJLH001 via transcriptomics and culturomics. Food Funct. 2019, 10, 3556–3566. [Google Scholar] [CrossRef] [PubMed]
  109. 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] [PubMed]
  110. Yan, F.; Li, N.; Shi, J.; Li, H.; Yue, Y.; Jiao, W.; Wang, N.; Song, Y.; Huo, G.; Li, B. Lactobacillus acidophilus alleviates type 2 diabetes by regulating hepatic glucose, lipid metabolism and gut microbiota in mice. Food Funct. 2019, 10, 5804–5815. [Google Scholar] [CrossRef]
  111. Bonfili, L.; Cecarini, V.; Gogoi, O.; Berardi, S.; Scarpona, S.; Angeletti, M.; Rossi, G.; Eleuteri, A.M. Gut microbiota manipulation through probiotics oral administration restores glucose homeostasis in a mouse model of Alzheimer’s disease. Neurobiol. Aging 2020, 87, 35–43. [Google Scholar] [CrossRef]
  112. Rodrigues, R.R.; Gurung, M.; Li, Z.; García-Jaramillo, M.; Greer, R.; Gaulke, C.; Bauchinger, F.; You, H.; Pederson, J.W.; Vasquez-Perez, S.; et al. Transkingdom interactions between Lactobacilli and hepatic mitochondria attenuate western diet-induced diabetes. Nat. Commun. 2021, 12, 101. [Google Scholar] [CrossRef]
  113. Wang, Y.; Wu, Y.; Wang, B.; Xu, H.; Mei, X.; Xu, X.; Zhang, X.; Ni, J.; Li, W. Bacillus amyloliquefaciens SC06 Protects Mice Against High-Fat Diet-Induced Obesity and Liver Injury via Regulating Host Metabolism and Gut Microbiota. Front. Microbiol. 2019, 10, 1161. [Google Scholar] [CrossRef]
  114. Park, K.-Y.; Kim, B.; Hyun, C.-K. Lactobacillus rhamnosus GG improves glucose tolerance through alleviating ER stress and suppressing macrophage activation in db/db mice. J. Clin. Biochem. Nutr. 2015, 56, 240–246. [Google Scholar] [CrossRef] [Green Version]
  115. Hsu, Y.-J.; Chiu, C.-C.; Lee, M.-C.; Huang, W.-C. Combination of Treadmill Aerobic Exercise with Bifidobacterium longum OLP-01 Supplementation for Treatment of High-Fat Diet-Induced Obese Murine Model. Obes. Facts 2021, 14, 306–319. [Google Scholar] [CrossRef]
  116. Kim, S.J.; Choi, S.I.; Jang, M.; Jeong, Y.A.; Kang, C.H.; Kim, G.H. Combination of Limosilactobacillus fermentum MG4231 and MG4244 attenuates lipid accumulation in high-fat diet-fed obese mice. Benef. Microbes 2021, 12, 479–491. [Google Scholar] [CrossRef]
  117. Bonfili, L.; Cecarini, V.; Berardi, S.; Scarpona, S.; Suchodolski, J.S.; Nasuti, C.; Fiorini, D.; Boarelli, M.C.; Rossi, G.; Eleuteri, A.M. Microbiota modulation counteracts Alzheimer’s disease progression influencing neuronal proteolysis and gut hormones plasma levels. Sci. Rep. 2017, 7, 2426. [Google Scholar] [CrossRef]
  118. Chen, P.; Zhang, Q.; Dang, H.; Liu, X.; Tian, F.; Zhao, J.; Chen, Y.; Zhang, H.; Chen, W. Oral administration of Lactobacillus rhamnosus CCFM0528 improves glucose tolerance and cytokine secretion in high-fat-fed, streptozotocin-induced type 2 diabetic mice. J. Funct. Foods 2014, 10, 318–326. [Google Scholar] [CrossRef]
  119. Komori, T. Functions of Osteocalcin in Bone, Pancreas, Testis, and Muscle. Int. J. Mol. Sci. 2020, 21, 7513. [Google Scholar] [CrossRef]
  120. Zoch, M.L.; Clemens, T.L.; Riddle, R.C. New insights into the biology of osteocalcin. Bone 2016, 82, 42–49. [Google Scholar] [CrossRef] [Green Version]
  121. Zanatta, L.C.B.; Boguszewski, C.L.; Borba, V.Z.C.; Kulak, C.A.M. Osteocalcin, energy and glucose metabolism. Arq. Bras. Endocrinol. Metabol. 2014, 58, 444–451. [Google Scholar] [CrossRef] [Green Version]
  122. Dang, F.; Jiang, Y.; Pan, R.; Zhou, Y.; Wu, S.; Wang, R.; Zhuang, K.; Zhang, W.; Li, T.; Man, C. Administration of Lactobacillus paracasei ameliorates type 2 diabetes in mice. Food Funct. 2018, 9, 3630–3639. [Google Scholar] [CrossRef]
  123. Stavropoulou, E.; Bezirtzoglou, E. Probiotics in Medicine: A Long Debate. Front. Immunol. 2020, 11, 2192. [Google Scholar] [CrossRef]
  124. Hylemon, P.B.; Takabe, K.; Dozmorov, M.; Nagahashi, M.; Zhou, H. Bile acids as global regulators of hepatic nutrient metabolism. Liver Res. 2017, 1, 10–16. [Google Scholar] [CrossRef]
  125. Martoni, C.J.; Labbé, A.; Ganopolsky, J.G.; Prakash, S.; Jones, M.L. Changes in bile acids, FGF-19 and sterol absorption in response to bile salt hydrolase active L. reuteri NCIMB 30242. Gut Microbes 2015, 6, 57–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Holt, J.A.; Luo, G.; Billin, A.N.; Bisi, J.; McNeill, Y.Y.; Kozarsky, K.F.; Donahee, M.; Wang, D.Y.; Mansfield, T.A.; Kliewer, S.A.; et al. Definition of a novel growth factor-dependent signal cascade for the suppression of bile acid biosynthesis. Genes Dev. 2003, 17, 1581–1591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Zhong, Z.; Zhang, W.; Du, R.; Meng, H.; Zhang, H. Lactobacillus casei Zhang stimulates lipid metabolism in hypercholesterolemic rats by affecting gene expression in the liver. Eur. J. Lipid Sci. Technol. 2012, 114, 244–252. [Google Scholar] [CrossRef]
  128. Zhang, Y.; Guo, X.; Guo, J.; He, Q.; Li, H.; Song, Y.; Zhang, H. Lactobacillus casei reduces susceptibility to type 2 diabetes via microbiota-mediated body chloride ion influx. Sci. Rep. 2014, 4, 5654. [Google Scholar] [CrossRef] [Green Version]
  129. Bleau, C.; Karelis, A.D.; St-Pierre, D.H.; Lamontagne, L. Crosstalk between intestinal microbiota, adipose tissue and skeletal muscle as an early event in systemic low-grade inflammation and the development of obesity and diabetes. Diabetes. Metab. Res. Rev. 2015, 31, 545–561. [Google Scholar] [CrossRef]
  130. Hotamisligil, G.S. Inflammation and metabolic disorders. Nature 2006, 444, 860–867. [Google Scholar] [CrossRef]
  131. 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]
  132. 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]
  133. Yoon, Y.; Kim, G.; Noh, M.; Park, J.; Jang, M.; Fang, S.; Park, H. Lactobacillus fermentum promotes adipose tissue oxidative phosphorylation to protect against diet-induced obesity. Exp. Mol. Med. 2020, 52, 1574–1586. [Google Scholar] [CrossRef]
  134. Wang, T.; Yan, H.; Lu, Y.; Li, X.; Wang, X.; Shan, Y.; Yi, Y.; Liu, B.; Zhou, Y.; Lü, X. Anti-obesity effect of Lactobacillus rhamnosus LS-8 and Lactobacillus crustorum MN047 on high-fat and high-fructose diet mice base on inflammatory response alleviation and gut microbiota regulation. Eur. J. Nutr. 2019, 59, 2709–2728. [Google Scholar] [CrossRef]
  135. Deng, L.; Ou, Z.; Huang, D.; Li, C.; Lu, Z.; Liu, W.; Wu, F.; Nong, C.; Gao, J.; Peng, Y. Diverse effects of different Akkermansia muciniphila genotypes on Brown adipose tissue inflammation and whitening in a high-fat-diet murine model. Microb. Pathog. 2020, 147, 104353. [Google Scholar] [CrossRef]
  136. Carvalho-Filho, M.A.; Ueno, M.; Hirabara, S.M.; Seabra, A.B.; Carvalheira, J.B.C.; de Oliveira, M.G.; Velloso, L.A.; Curi, R.; Saad, M.J.A. S-Nitrosation of the Insulin Receptor, Insulin Receptor Substrate 1, and Protein Kinase B/Akt: A Novel Mechanism of Insulin Resistance. Diabetes 2005, 54, 959–967. [Google Scholar] [CrossRef] [Green Version]
  137. Fang, H.; Judd, R.L. Adiponectin Regulation and Function. Compr. Physiol. 2018, 8, 1031–1063. [Google Scholar] [CrossRef]
  138. Le, T.K.C.; Hosaka, T.; Nguyen, T.T.; Kassu, A.; Dang, T.O.; Tran, H.B.; Pham, T.P.; Tran, Q.B.; Le, T.H.H.; Pham, X. Da Bifidobacterium species lower serum glucose, increase expressions of insulin signaling proteins, and improve adipokine profile in diabetic mice. Biomed. Res. 2015, 36, 63–70. [Google Scholar] [CrossRef] [Green Version]
  139. Liu, Q.; Liu, Y.; Li, F.; Gu, Z.; Liu, M.; Shao, T.; Zhang, L.; Zhou, G.; Pan, C.; He, L.; et al. Probiotic culture supernatant improves metabolic function through FGF21-adiponectin pathway in mice. J. Nutr. Biochem. 2020, 75, 108256. [Google Scholar] [CrossRef]
  140. Tyagi, S.; Gupta, P.; Saini, A.S.; Kaushal, C.; Sharma, S. The peroxisome proliferator-activated receptor: A family of nuclear receptors role in various diseases. J. Adv. Pharm. Technol. Res. 2011, 2, 236–240. [Google Scholar] [CrossRef]
  141. Molina-Tijeras, J.A.; Diez-Echave, P.; Vezza, T.; Hidalgo-García, L.; Ruiz-Malagón, A.J.; Rodríguez-Sojo, M.J.; Romero, M.; Robles-Vera, I.; García, F.; Plaza-Diaz, J.; et al. Lactobacillus fermentum CECT5716 ameliorates high fat diet-induced obesity in mice through modulation of gut microbiota dysbiosis. Pharmacol. Res. 2021, 167, 105471. [Google Scholar] [CrossRef]
  142. Kwon, J.; Kim, B.; Lee, C.; Joung, H.; Kim, B.K.; Choi, I.S.; Hyun, C.K. Comprehensive amelioration of high-fat diet-induced metabolic dysfunctions through activation of the PGC-1α pathway by probiotics treatment in mice. PLoS ONE 2020, 15, e0228932. [Google Scholar] [CrossRef] [Green Version]
  143. López-Bermejo, A.; Botas-Cervero, P.; Ortega-Delgado, F.; Delgado, E.; García-Gil, M.M.; Funahashi, T.; Ricart, W.; Fernández-Real, J.M. Association of ADIPOR2 With Liver Function Tests in Type 2 Diabetic Subjects. Obesity 2008, 16, 2308–2313. [Google Scholar] [CrossRef]
  144. Olefsky, J.M.; Saltiel, A.R. PPAR gamma and the treatment of insulin resistance. Trends Endocrinol. Metab. 2000, 11, 362–368. [Google Scholar] [CrossRef]
  145. Mueckler, M.; Thorens, B. The SLC2 (GLUT) family of membrane transporters. Mol. Aspects Med. 2013, 34, 121–138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Li, C.; Ding, Q.; Nie, S.-P.; Zhang, Y.-S.; Xiong, T.; Xie, M.-Y. Carrot Juice Fermented with Lactobacillus plantarum NCU116 Ameliorates Type 2 Diabetes in Rats. J. Agric. Food Chem. 2014, 62, 11884–11891. [Google Scholar] [CrossRef] [PubMed]
  147. Zhang, Z.; Liang, X.; Lv, Y.; Yi, H.; Chen, Y.; Bai, L.; Zhou, H.; Liu, T.; Li, R.; Zhang, L. Evaluation of probiotics for improving and regulation metabolism relevant to type 2 diabetes in vitro. J. Funct. Foods 2020, 64, 103664. [Google Scholar] [CrossRef]
  148. Yadav, H.; Quijano, C.; Kamaraju, A.K.; Gavrilova, O.; Malek, R.; Chen, W.; Zerfas, P.; Zhigang, D.; Wright, E.C.; Stuelten, C.; et al. Protection from obesity and diabetes by blockade of TGF-β/Smad3 signaling. Cell Metab. 2011, 14, 67–79. [Google Scholar] [CrossRef] [Green Version]
  149. Sousa-Pinto, B.; Gonçalves, L.; Rodrigues, A.R.; Tomada, I.; Almeida, H.; Neves, D.; Gouveia, A.M. Characterization of TGF-β expression and signaling profile in the adipose tissue of rats fed with high-fat and energy-restricted diets. J. Nutr. Biochem. 2016, 38, 107–115. [Google Scholar] [CrossRef]
  150. 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]
  151. Flamment, M.; Hajduch, E.; Ferré, P.; Foufelle, F. New insights into ER stress-induced insulin resistance. Trends Endocrinol. Metab. 2012, 23, 381–390. [Google Scholar] [CrossRef]
  152. Kim, S.-H.; Huh, C.-S.; Choi, I.-D.; Jeong, J.-W.; Ku, H.-K.; Ra, J.-H.; Kim, T.-Y.; Kim, G.-B.; Sim, J.-H.; Ahn, Y.-T. The anti-diabetic activity of Bifidobacterium lactis HY8101 in vitro and in vivo. J. Appl. Microbiol. 2014, 117, 834–845. [Google Scholar] [CrossRef]
  153. Kim, S.-W.; Park, K.-Y.; Kim, B.; Kim, E.; Hyun, C.-K. Lactobacillus rhamnosus GG improves insulin sensitivity and reduces adiposity in high-fat diet-fed mice through enhancement of adiponectin production. Biochem. Biophys. Res. Commun. 2013, 431, 258–263. [Google Scholar] [CrossRef]
  154. Oberley, L.W. Free radicals and diabetes. Free Radic. Biol. Med. 1988, 5, 113–124. [Google Scholar] [CrossRef]
  155. Wang, G.; Li, X.; Zhao, J.; Zhang, H.; Chen, W. Lactobacillus casei CCFM419 attenuates type 2 diabetes via a gut microbiota dependent mechanism. Food Funct. 2017, 8, 3155–3164. [Google Scholar] [CrossRef]
  156. Calcinaro, F.; Dionisi, S.; Marinaro, M.; Candeloro, P.; Bonato, V.; Marzotti, S.; Corneli, R.B.; Ferretti, E.; Gulino, A.; Grasso, F.; et al. Oral probiotic administration induces interleukin-10 production and prevents spontaneous autoimmune diabetes in the non-obese diabetic mouse. Diabetologia 2005, 48, 1565–1575. [Google Scholar] [CrossRef] [Green Version]
  157. Heeney, D.D.; Zhai, Z.; Bendiks, Z.; Barouei, J.; Martinic, A.; Slupsky, C.; Marco, M.L. Lactobacillus plantarum bacteriocin is associated with intestinal and systemic improvements in diet-induced obese mice and maintains epithelial barrier integrity in vitro. Gut Microbes 2018, 10, 382–397. [Google Scholar] [CrossRef] [Green Version]
  158. Huang, D.; Gao, J.; Li, C.; Nong, C.; Huang, W.; Zheng, X.; Li, S.; Peng, Y. A potential probiotic bacterium for antipsychotic-induced metabolic syndrome: Mechanisms underpinning how Akkermansia muciniphila subtype improves olanzapine-induced glucose homeostasis in mice. Psychopharmacology 2021, 238, 2543–2553. [Google Scholar] [CrossRef]
  159. Park, S.-S.; Yang, G.; Kim, E. Lactobacillus acidophilus NS1 Reduces Phosphoenolpyruvate Carboxylase Expression by Regulating HNF4α Transcriptional Activity. Korean J. Food Sci. Anim. Resour. 2017, 37, 529–534. [Google Scholar] [CrossRef] [Green Version]
  160. Walesky, C.; Apte, U. Role of hepatocyte nuclear factor 4α (HNF4α) in cell proliferation and cancer. Gene Expr. 2015, 16, 101–108. [Google Scholar] [CrossRef] [Green Version]
  161. Li, Y.; Wang, L.; Zhou, L.; Song, Y.; Ma, S.; Yu, C.; Zhao, J.; Xu, C.; Gao, L. Thyroid stimulating hormone increases hepatic gluconeogenesis via CRTC2. Mol. Cell. Endocrinol. 2017, 446, 70–80. [Google Scholar] [CrossRef]
  162. Liu, Y.; Yang, L.; Zhang, Y.; Liu, X.; Wu, Z.; Gilbert, R.G.; Deng, B.; Wang, K. Dendrobium officinale polysaccharide ameliorates diabetic hepatic glucose metabolism via glucagon-mediated signaling pathways and modifying liver-glycogen structure. J. Ethnopharmacol. 2020, 248, 112308. [Google Scholar] [CrossRef]
  163. Fountain, J.H.; Lappin, S.L. Physiology, Renin Angiotensin System; StatPearls: Treasure Island, FL, USA, 2021. [Google Scholar]
  164. Xu, J.; Fan, J.; Wu, F.; Huang, Q.; Guo, M.; Lv, Z.; Han, J.; Duan, L.; Hu, G.; Chen, L.; et al. The ACE2/angiotensin-(1-7)/Mas receptor axis: Pleiotropic roles in cancer. Front. Physiol. 2017, 8, 276. [Google Scholar] [CrossRef]
  165. Santos, S.H.S.; Fernandes, L.R.; Mario, É.G.; Ferreira, A.V.M.; Pôrto, L.C.J.; Alvarez-Leite, J.I.; Botion, L.M.; Bader, M.; Alenina, N.; Santos, R.A.S. Mas Deficiency in FVB/N Mice Produces Marked Changes in Lipid and Glycemic Metabolism. Diabetes 2008, 57, 340–347. [Google Scholar] [CrossRef] [Green Version]
  166. Bilman, V.; Mares-Guia, L.; Nadu, A.P.; Bader, M.; Campagnole-Santos, M.J.; Santos, R.A.S.; Santos, S.H.S. Decreased hepatic gluconeogenesis in transgenic rats with increased circulating angiotensin-(1-7). Peptides 2012, 37, 247–251. [Google Scholar] [CrossRef] [Green Version]
  167. Li, X.; Xu, Q.; Jiang, T.; Fang, S.; Wang, G.; Zhao, J.; Zhang, H.; Chen, W. A comparative study of the antidiabetic effects exerted by live and dead multi-strain probiotics in the type 2 diabetes model of mice. Food Funct. 2016, 7, 4851–4860. [Google Scholar] [CrossRef]
  168. 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]
  169. Wright, E.M.; Loo, D.D.F.; Hirayama, B.A. Biology of Human Sodium Glucose Transporters. Physiol. Rev. 2011, 91, 733–794. [Google Scholar] [CrossRef] [Green Version]
  170. Korkmaz, O.A.; Sumlu, E.; Koca, H.B.; Pektas, M.B.; Kocabas, A.; Sadi, G.; Akar, F.; Korkmaz, O.A.; Sumlu, E.; Koca, H.B.; et al. Effects of Lactobacillus Plantarum and Lactobacillus Helveticus on Renal Insulin Signaling, Inflammatory Markers, and Glucose Transporters in High-Fructose-Fed Rats. Medicina 2019, 55, 207. [Google Scholar] [CrossRef] [Green Version]
  171. Eckburg, P.B.; Bik, E.M.; Bernstein, C.N.; Purdom, E.; Dethlefsen, L.; Sargent, M.; Gill, S.R.; Nelson, K.E.; Relman, D.A. Diversity of the human intestinal microbial flora. Science 2005, 308, 1635–1638. [Google Scholar] [CrossRef] [Green Version]
  172. Voreades, N.; Kozil, A.; Weir, T.L. Diet and the development of the human intestinal microbiome. Front. Microbiol. 2014, 5, 494. [Google Scholar] [CrossRef] [Green Version]
  173. 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] [Green Version]
  174. Stenvinkel, P. Obesity-a disease with many aetiologies disguised in the same oversized phenotype: Has the overeating theory failed? Nephrol. Dial. Transplant 2014, 30, 1656–1664. [Google Scholar] [CrossRef] [Green Version]
  175. Kallus, S.J.; Brandt, L.J. The intestinal microbiota and obesity. J. Clin. Gastroenterol. 2012, 46, 16–24. [Google Scholar] [CrossRef]
  176. 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]
  177. Ley, R.E.; Turnbaugh, P.J.; Klein, S.; Gordon, J.I. Microbial ecology: Human gut microbes associated with obesity. Nature 2006, 444, 1022–1023. [Google Scholar] [CrossRef] [PubMed]
  178. Armougom, F.; Henry, M.; Vialettes, B.; Raccah, D.; Raoult, D. Monitoring bacterial community of human gut microbiota reveals an increase in Lactobacillus in obese patients and Methanogens in anorexic patients. PLoS ONE 2009, 4, e7125. [Google Scholar] [CrossRef] [PubMed]
  179. Million, M.; Maraninchi, M.; Henry, M.; Armougom, F.; Richet, H.; Carrieri, P.; Valero, R.; Raccah, D.; Vialettes, B.; Raoult, D. Obesity-associated gut microbiota is enriched in Lactobacillus reuteri and depleted in Bifidobacterium animalis and Methanobrevibacter smithii. Int. J. Obes. 2012, 36, 817–825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Collado, M.C.; Isolauri, E.; Laitinen, K.; Salminen, S. Distinct composition of gut microbiota during pregnancy in overweight and normal-weight women. Am. J. Clin. Nutr. 2008, 88, 894–899. [Google Scholar] [CrossRef] [PubMed]
  181. Schwiertz, A.; Taras, D.; Schäfer, K.; Beijer, S.; Bos, N.A.; Donus, C.; Hardt, P.D. Microbiota and SCFA in lean and overweight healthy subjects. Obesity 2010, 18, 190–195. [Google Scholar] [CrossRef] [PubMed]
  182. Mai, V.; McCrary, Q.M.; Sinha, R.; Glei, M. Associations between dietary habits and body mass index with gut microbiota composition and fecal water genotoxicity: An observational study in African American and Caucasian American volunteers. Nutr. J. 2009, 8, 49. [Google Scholar] [CrossRef] [Green Version]
  183. Arumugam, M.; Raes, J.; Pelletier, E.; Le Paslier, D.; Yamada, T.; Mende, D.R.; Fernandes, G.R.; Tap, J.; Bruls, T.; Batto, J.-M.; et al. Enterotypes of the human gut microbiome. Nature 2011, 473, 174–180. [Google Scholar] [CrossRef]
  184. Million, M.; Lagier, J.-C.; Yahav, D.; Paul, M. Gut bacterial microbiota and obesity. Clin. Microbiol. Infect. 2013, 19, 305–313. [Google Scholar] [CrossRef] [Green Version]
  185. Larsen, N.; Vogensen, F.K.; van den Berg, F.W.J.; Nielsen, D.S.; Andreasen, A.S.; Pedersen, B.K.; Al-Soud, W.A.; Sørensen, S.J.; Hansen, L.H.; Jakobsen, M. Gut Microbiota in Human Adults with Type 2 Diabetes Differs from Non-Diabetic Adults. PLoS ONE 2010, 5, e9085. [Google Scholar] [CrossRef]
  186. Ivey, K.L.; Hodgson, J.M.; Kerr, D.A.; Lewis, J.R.; Thompson, P.L.; Prince, R.L. The effects of probiotic bacteria on glycaemic control in overweight men and women: A randomised controlled trial. Eur. J. Clin. Nutr. 2014, 68, 447–452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Solito, A.; Bozzi Cionci, N.; Calgaro, M.; Caputo, M.; Vannini, L.; Hasballa, I.; Archero, F.; Giglione, E.; Ricotti, R.; Walker, G.E.; et al. Supplementation with Bifidobacterium breve BR03 and B632 strains improved insulin sensitivity in children and adolescents with obesity in a cross-over, randomized double-blind placebo-controlled trial. Clin. Nutr. 2021, 40, 4585–4594. [Google Scholar] [CrossRef] [PubMed]
  188. Zavratnik, A.; Krajnc, M. Analysis of pregnant women with the diagnosis of gestational diabetes mellitus without oral glucose tolerance test confirmation in early pregnancy. Acta Med. -Biotech. 2018, 11, 27–36. [Google Scholar]
  189. Laitinen, K.; Poussa, T.; Isolauri, E.; Nutrition, Allergy, Mucosal Immunology and Intestinal Microbiota Group. Probiotics and dietary counselling contribute to glucose regulation during and after pregnancy: A randomised controlled trial. Br. J. Nutr. 2008, 101, 1679–1687. [Google Scholar] [CrossRef] [Green Version]
  190. Dallanora, S.; Medeiros de Souza, Y.; Deon, R.G.; Tracey, C.A.; Freitas-Vilela, A.A.; Wurdig Roesch, L.F.; Hack Mendes, R. Do probiotics effectively ameliorate glycemic control during gestational diabetes? A systematic review. Arch. Gynecol. Obstet. 2018, 298, 477–485. [Google Scholar] [CrossRef]
  191. Karamali, M.; Dadkhah, F.; Sadrkhanlou, M.; Jamilian, M.; Ahmadi, S.; Tajabadi-Ebrahimi, M.; Jafari, P.; Asemi, Z. Effects of probiotic supplementation on glycaemic control and lipid profiles in gestational diabetes: A randomized, double-blind, placebo-controlled trial. Diabetes Metab. 2016, 42, 234–241. [Google Scholar] [CrossRef]
  192. Wickens, K.L.; Barthow, C.A.; Murphy, R.; Abels, P.R.; Maude, R.M.; Stone, P.R.; Mitchell, E.A.; Stanley, T.V.; Purdie, G.L.; Kang, J.M.; et al. Early pregnancy probiotic supplementation with Lactobacillus rhamnosus HN001 may reduce the prevalence of gestational diabetes mellitus: A randomised controlled trial. Br. J. Nutr. 2017, 117, 804–813. [Google Scholar] [CrossRef] [Green Version]
  193. Dolatkhah, N.; Hajifaraji, M.; Abbasalizadeh, F.; Aghamohammadzadeh, N.; Mehrabi, Y.; Mesgari Abbasi, M. Is there a value for probiotic supplements in gestational diabetes mellitus? A randomized clinical trial. J. Health Popul. Nutr. 2015, 33, 25. [Google Scholar] [CrossRef] [Green Version]
  194. Sahhaf Ebrahimi, F.; Homayouni Rad, A.; Mosen, M.; Abbasalizadeh, F.; Tabrizi, A.; Khalili, L. Effect of L. acidophilus and B. lactis on blood glucose in women with gestational diabetes mellitus: A randomized placebo-controlled trial. Diabetol. Metab. Syndr. 2019, 11, 75. [Google Scholar] [CrossRef]
  195. Shahriari, A.; Karimi, E.; Shahriari, M.; Aslani, N.; Khooshideh, M.; Arab, A. The effect of probiotic supplementation on the risk of gestational diabetes mellitus among high-risk pregnant women: A parallel double-blind, randomized, placebo-controlled clinical trial. Biomed. Pharmacother. 2021, 141, 111915. [Google Scholar] [CrossRef]
  196. Tsigalou, C.; Konstantinidis, T.; Stavropoulou, E.; Bezirtzoglou, E.E.; Tsakris, A. Potential Elimination of Human Gut Resistome by Exploiting the Benefits of Functional Foods. Front. Microbiol. 2020, 11, 50. [Google Scholar] [CrossRef] [Green Version]
  197. Li, X.; Huang, Y.; Song, L.; Xiao, Y.; Lu, S.; Xu, J.; Li, J.; Ren, Z. Lactobacillus plantarum prevents obesity via modulation of gut microbiota and metabolites in high-fat feeding mice. J. Funct. Foods 2020, 73, 104103. [Google Scholar] [CrossRef]
  198. 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]
  199. Li, K.K.; Tian, P.J.; Wang, S.D.; Lei, P.; Qu, L.; Huang, J.P.; Shan, Y.J.; Li, B. long Targeting gut microbiota: Lactobacillus alleviated type 2 diabetes via inhibiting LPS secretion and activating GPR43 pathway. J. Funct. Foods 2017, 38, 561–570. [Google Scholar] [CrossRef]
  200. Luo, J.; Zhang, H.; Lu, J.; Ma, C.L.; Chen, T. Antidiabetic effect of an engineered bacterium Lactobacillus plantarum-pMG36e -GLP-1 in monkey model. Synth. Syst. Biotechnol. 2021, 6, 272–282. [Google Scholar] [CrossRef]
  201. Zheng, Q.-X.; Jiang, X.-M.; Wang, H.-W.; Ge, L.; Lai, Y.-T.; Jiang, X.-Y.; Chen, F.; Huang, P.-P. Probiotic supplements alleviate gestational diabetes mellitus by restoring the diversity of gut microbiota: A study based on 16S rRNA sequencing. J. Microbiol. 2021, 59, 827–839. [Google Scholar] [CrossRef]
  202. Den Besten, G.; van Eunen, K.; Groen, A.K.; Venema, K.; Reijngoud, D.-J.; Bakker, B.M. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J. Lipid Res. 2013, 54, 2325–2340. [Google Scholar] [CrossRef] [Green Version]
  203. Martin-Gallausiaux, C.; Marinelli, L.; Blottière, H.M.; Larraufie, P.; Lapaque, N. SCFA: Mechanisms and functional importance in the gut. Proc. Nutr. Soc. 2021, 80, 37–49. [Google Scholar] [CrossRef]
  204. Primec, M.; Mičetić-Turk, D.; Langerholc, T. Analysis of short-chain fatty acids in human feces: A scoping review. Anal. Biochem. 2017, 526, 9–21. [Google Scholar] [CrossRef]
  205. Yadav, H.; Lee, J.-H.; Lloyd, J.; Walter, P.; Rane, S.G. Beneficial Metabolic Effects of a Probiotic via Butyrate-induced GLP-1 Hormone Secretion. J. Biol. Chem. 2013, 288, 25088–25097. [Google Scholar] [CrossRef] [Green Version]
  206. Troy, S.; Soty, M.; Ribeiro, L.; Laval, L.; Migrenne, S.; Fioramonti, X.; Pillot, B.; Fauveau, V.; Aubert, R.; Viollet, B.; et al. Intestinal Gluconeogenesis Is a Key Factor for Early Metabolic Changes after Gastric Bypass but Not after Gastric Lap-Band in Mice. Cell Metab. 2008, 8, 201–211. [Google Scholar] [CrossRef] [Green Version]
  207. Le Poul, E.; Loison, C.; Struyf, S.; Springael, J.Y.; Lannoy, V.; Decobecq, M.E.; Brezillon, S.; Dupriez, V.; Vassart, G.; Van Damme, J.; et al. Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J. Biol. Chem. 2003, 278, 25481–25489. [Google Scholar] [CrossRef] [Green Version]
  208. Horiuchi, H.; Kamikado, K.; Aoki, R.; Suganuma, N.; Nishijima, T.; Nakatani, A.; Kimura, I. Bifidobacterium animalis subsp. lactis GCL2505 modulates host energy metabolism via the short-chain fatty acid receptor GPR43. Sci. Rep. 2020, 10, 4158. [Google Scholar] [CrossRef] [Green Version]
  209. Lu, X.; Qiao, S.; Peng, C.; Yan, W.; Xu, Z.; Qu, J.; Hou, Y.; Zhao, S.; Chen, P.; Wang, T. Bornlisy Attenuates Colitis-Associated Colorectal Cancer via Inhibiting GPR43-Mediated Glycolysis. Front. Nutr. 2021, 8, 835. [Google Scholar] [CrossRef]
  210. Drucker, D.J. Mechanisms of Action and Therapeutic Application of Glucagon-like Peptide-1. Cell Metab. 2018, 27, 740–756. [Google Scholar] [CrossRef] [Green Version]
  211. Nauck, M.A.; Meier, J.J. Incretin hormones: Their role in health and disease. Diabetes Obes. Metab. 2018, 20, 5–21. [Google Scholar] [CrossRef]
  212. Simon, M.-C.; Strassburger, K.; Nowotny, B.; Kolb, H.; Nowotny, P.; Burkart, V.; Zivehe, F.; Hwang, J.-H.; Stehle, P.; Pacini, G.; et al. Intake of Lactobacillus reuteri Improves Incretin and Insulin Secretion in Glucose-Tolerant Humans: A Proof of Concept. Diabetes Care 2015, 38, 1827–1834. [Google Scholar] [CrossRef] [Green Version]
  213. Bjerg, A.T.; Kristensen, M.; Ritz, C.; Holst, J.J.; Rasmussen, C.; Leser, T.D.; Wellejus, A.; Astrup, A. Lactobacillus paracasei subsp paracasei L. casei W8 suppresses energy intake acutely. Appetite 2014, 82, 111–118. [Google Scholar] [CrossRef] [PubMed]
  214. Yadav, H.; Jain, S.; Sinha, P.R. Oral administration of dahi containing probiotic Lactobacillus acidophilus and Lactobacillus casei delayed the progression of streptozotocin-induced diabetes in rats. J. Dairy Res. 2008, 75, 189–195. [Google Scholar] [CrossRef] [PubMed]
  215. Yamano, T.; Tanida, M.; Niijima, A.; Maeda, K.; Okumura, N.; Fukushima, Y.; Nagai, K. Effects of the probiotic strain Lactobacillus johnsonii strain La1 on autonomic nerves and blood glucose in rats. Life Sci. 2006, 79, 1963–1967. [Google Scholar] [CrossRef] [PubMed]
  216. Zhu, Y.; Liu, Q.; Zhou, Z.; Ikeda, Y. PDX1, Neurogenin-3, and MAFA: Critical transcription regulators for beta cell development and regeneration. Stem Cell Res. Ther. 2017, 8, 240. [Google Scholar] [CrossRef] [Green Version]
  217. Duan, F.; Curtis, K.L.; March, J.C. Secretion of Insulinotropic Proteins by Commensal Bacteria: Rewiring the Gut To Treat Diabetes. Appl. Environ. Microbiol. 2008, 74, 7437–7438. [Google Scholar] [CrossRef] [Green Version]
  218. Belguesmia, Y.; Alard, J.; Mendil, R.; Ravallec, R.; Grangette, C.; Drider, D.; Cudennec, B. In vitro probiotic properties of selected lactobacilli and multi-strain consortium on immune function, gut barrier strengthening and gut hormone secretion. J. Funct. Foods 2019, 57, 382–391. [Google Scholar] [CrossRef]
  219. Hung, S.-C.; Tseng, W.-T.; Pan, T.-M. Lactobacillus paracasei subsp. paracasei NTU 101 ameliorates impaired glucose tolerance induced by a high-fat, high-fructose diet in Sprague-Dawley rats. J. Funct. Foods 2016, 24, 472–481. [Google Scholar] [CrossRef]
  220. Tian, P.; Li, B.; He, C.; Song, W.; Hou, A.; Tian, S.; Meng, X.; Li, K.; Shan, Y. Antidiabetic (type 2) effects of Lactobacillus G15 and Q14 in rats through regulation of intestinal permeability and microbiota. Food Funct. 2016, 7, 3789–3797. [Google Scholar] [CrossRef]
  221. Rooj, A.K.; Kimura, Y.; Buddington, R.K. Metabolites produced by probiotic Lactobacilli rapidly increase glucose uptake by Caco-2 cells. BMC Microbiol. 2010, 10, 16. [Google Scholar] [CrossRef] [Green Version]
  222. Primec, M.; Škorjanc, D.; Langerholc, T.; Mičetić-Turk, D.; Gorenjak, M. Specific Lactobacillus probiotic strains decrease transepithelial glucose transport through GLUT2 downregulation in intestinal epithelial cell models. Nutr. Res. 2021, 86, 10–22. [Google Scholar] [CrossRef]
  223. Kolb, H.; Kempf, K.; Röhling, M.; Lenzen-Schulte, M.; Schloot, N.C.; Martin, S. Ketone bodies: From enemy to friend and guardian angel. BMC Med. 2021, 19, 313. [Google Scholar] [CrossRef]
  224. Liao, C.A.; Huang, C.H.; Ho, H.H.; Chen, J.F.; Kuo, Y.W.; Lin, J.H.; Tsai, S.Y.; Tsai, H.Y.; Yeh, Y.T. A Combined Supplement of Probiotic Strains AP-32, bv-77, and CP-9 Increased Akkermansia mucinphila and Reduced Non-Esterified Fatty Acids and Energy Metabolism in HFD-Induced Obese Rats. Nutrients 2022, 14, 527. [Google Scholar] [CrossRef]
  225. Zhang, Z.; Mocanu, V.; Cai, C.; Dang, J.; Slater, L.; Deehan, E.C.; Walter, J.; Madsen, K.L. Impact of Fecal Microbiota Transplantation on Obesity and Metabolic Syndrome-A Systematic Review. Nutrients 2019, 11, 2291. [Google Scholar] [CrossRef] [Green Version]
  226. Aron-Wisnewsky, J.; Clément, K.; Nieuwdorp, M. Fecal Microbiota Transplantation: A Future Therapeutic Option for Obesity/Diabetes? Curr. Diab. Rep. 2019, 19, 51. [Google Scholar] [CrossRef] [PubMed]
  227. Huda, M.N.; Kim, M.; Bennett, B.J. Modulating the Microbiota as a Therapeutic Intervention for Type 2 Diabetes. Front. Endocrinol. 2021, 12, 632335. [Google Scholar] [CrossRef] [PubMed]
  228. Allegretti, J.R.; Kassam, Z.; Hurtado, J.; Marchesi, J.R.; Mullish, B.H.; Chiang, A.; Thompson, C.C.; Cummings, B.P. Impact of fecal microbiota transplantation with capsules on the prevention of metabolic syndrome among patients with obesity. Hormones 2021, 20, 209–211. [Google Scholar] [CrossRef] [PubMed]
  229. Proença, I.M.; Allegretti, J.R.; Bernardo, W.M.; de Moura, D.T.H.; Ponte Neto, A.M.; Matsubayashi, C.O.; Flor, M.M.; Kotinda, A.P.S.T.; de Moura, E.G.H. Fecal microbiota transplantation improves metabolic syndrome parameters: Systematic review with meta-analysis based on randomized clinical trials. Nutr. Res. 2020, 83, 1–14. [Google Scholar] [CrossRef]
Figure 1. Effect of probiotics on adipose tissue. TNF-α: tumour necrosis factor alpha; TGF-β: transforming growth factor beta; IL-6: interleukin-6; IL-1β: interleukin 1 beta; AdipoR2; adiponectin receptor 2; TGF-β: transforming growth factor beta; SIRT1: sirtuin (silent mating type information regulation 2 homolog) 1; PPARα: peroxisome proliferator-activated receptor α; PPAR-γ: peroxisome proliferator-activated receptor gamma; FGF21: fibroblast growth factor 21; GLUT4: glucose transporter 4; ↓: decrease; ↑: increase.
Figure 1. Effect of probiotics on adipose tissue. TNF-α: tumour necrosis factor alpha; TGF-β: transforming growth factor beta; IL-6: interleukin-6; IL-1β: interleukin 1 beta; AdipoR2; adiponectin receptor 2; TGF-β: transforming growth factor beta; SIRT1: sirtuin (silent mating type information regulation 2 homolog) 1; PPARα: peroxisome proliferator-activated receptor α; PPAR-γ: peroxisome proliferator-activated receptor gamma; FGF21: fibroblast growth factor 21; GLUT4: glucose transporter 4; ↓: decrease; ↑: increase.
Life 12 01187 g001
Figure 2. Effect of probiotics on skeletal muscles. GSK-3β: glycogen synthase kinase 3 beta; PI3K/AKT: phosphoinositide 3-kinase/phosphorylated protein kinase B signalling pathway; PPAR-γ: peroxisome proliferator-activated receptor gamma; GLUT4: glucose transporter 4; AMPK: 5′ adenosine monophosphate-activated protein kinase; ↓: decrease; ↑: increase.
Figure 2. Effect of probiotics on skeletal muscles. GSK-3β: glycogen synthase kinase 3 beta; PI3K/AKT: phosphoinositide 3-kinase/phosphorylated protein kinase B signalling pathway; PPAR-γ: peroxisome proliferator-activated receptor gamma; GLUT4: glucose transporter 4; AMPK: 5′ adenosine monophosphate-activated protein kinase; ↓: decrease; ↑: increase.
Life 12 01187 g002
Figure 3. Effect of probiotics on liver. Keap1/Nrf2: Kelch-like ECH-associated protein 1/Nuclear factor-erythroid factor 2-related factor 2; Gnas: Gs protein alpha-subunit; PKA: cAmp-dependent protein kinase; CRTC2: CREB-regulated transcriptional coactivator 2; PEPCK: phosphoenolpyruvate carboxykinase; G6PC: glucose-6-phosphatase catalytic subunit; ACE2/Ang-(1–7)/MasR): angiotensin-converting enzyme 2/angiotensin-(1–7)/mitochondrial assembly receptor; GSK-3β: glycogen synthase kinase 3 beta; FOXO1: Forehead Box O1; GS: glycogen synthase; PFK-1: phosphofructokinase-1; TNF-α: tumour necrosis factor alpha; IL-6: interleukin-6; IL-1β: interleukin 1 beta; PI3K/AKT: phosphoinositide 3-kinase/phosphorylated protein kinase B signalling pathway; PCK1: phosphoenolpyruvate carboxykinase 1; GLUT2: glucose transporter 2; ↓: decrease; ↑: increase.
Figure 3. Effect of probiotics on liver. Keap1/Nrf2: Kelch-like ECH-associated protein 1/Nuclear factor-erythroid factor 2-related factor 2; Gnas: Gs protein alpha-subunit; PKA: cAmp-dependent protein kinase; CRTC2: CREB-regulated transcriptional coactivator 2; PEPCK: phosphoenolpyruvate carboxykinase; G6PC: glucose-6-phosphatase catalytic subunit; ACE2/Ang-(1–7)/MasR): angiotensin-converting enzyme 2/angiotensin-(1–7)/mitochondrial assembly receptor; GSK-3β: glycogen synthase kinase 3 beta; FOXO1: Forehead Box O1; GS: glycogen synthase; PFK-1: phosphofructokinase-1; TNF-α: tumour necrosis factor alpha; IL-6: interleukin-6; IL-1β: interleukin 1 beta; PI3K/AKT: phosphoinositide 3-kinase/phosphorylated protein kinase B signalling pathway; PCK1: phosphoenolpyruvate carboxykinase 1; GLUT2: glucose transporter 2; ↓: decrease; ↑: increase.
Life 12 01187 g003
Figure 4. Effect of probiotics on kidney and pancreas. IL-6: interleukin-6; IRS-1: insulin receptor substrate 1 protein; eNOS: endothelial nitric oxide synthase; AKT: protein kinase B; SGLT2: sodium-glucose cotransporter 2; p65: transcription nuclear factor NF-kappa-B p65 subunit; TNF-α: tumour necrosis factor alpha; TLR4: toll-like receptor 4; IL-1β: interleukin 1 beta; IL-10: interleukin 10; PDX-1: insulin promoter factor 1; ClC-2: chloride channel protein 2; ↓: decrease; ↑: increase.
Figure 4. Effect of probiotics on kidney and pancreas. IL-6: interleukin-6; IRS-1: insulin receptor substrate 1 protein; eNOS: endothelial nitric oxide synthase; AKT: protein kinase B; SGLT2: sodium-glucose cotransporter 2; p65: transcription nuclear factor NF-kappa-B p65 subunit; TNF-α: tumour necrosis factor alpha; TLR4: toll-like receptor 4; IL-1β: interleukin 1 beta; IL-10: interleukin 10; PDX-1: insulin promoter factor 1; ClC-2: chloride channel protein 2; ↓: decrease; ↑: increase.
Life 12 01187 g004
Figure 5. Effect of probiotics on intestine. SCFA: short-chain fatty acids; PI3K/AKT: phosphoinositide 3-kinase/phosphorylated protein kinase B signalling pathway; GPR43/41: G protein-coupled receptor 43/41; GLP-1: glucagon-like polypeptide-1; TLR4: toll-like receptor 4; TLR2: toll-like receptor 2; TNF-α: tumour necrosis factor alpha; IL-6: interleukin-6; GLUT2: glucose transporter 2; GLUT5: glucose transporter 5; SGLT1: sodium-glucose cotransporter 1; ↓: decrease; ↑: increase; *, ", ^, °, #: each symbol stands for a different probiotic effect, which has a corresponding effect on glucose metabolism and homeostasis.
Figure 5. Effect of probiotics on intestine. SCFA: short-chain fatty acids; PI3K/AKT: phosphoinositide 3-kinase/phosphorylated protein kinase B signalling pathway; GPR43/41: G protein-coupled receptor 43/41; GLP-1: glucagon-like polypeptide-1; TLR4: toll-like receptor 4; TLR2: toll-like receptor 2; TNF-α: tumour necrosis factor alpha; IL-6: interleukin-6; GLUT2: glucose transporter 2; GLUT5: glucose transporter 5; SGLT1: sodium-glucose cotransporter 1; ↓: decrease; ↑: increase; *, ", ^, °, #: each symbol stands for a different probiotic effect, which has a corresponding effect on glucose metabolism and homeostasis.
Life 12 01187 g005
Table 1. Positive probiotic effects on some blood parameters in selected in vivo human studies.
Table 1. Positive probiotic effects on some blood parameters in selected in vivo human studies.
Altered Blood ParametersType of ProbioticPhysiological/
Pathological Condition
Reference
↓ Postprandial blood glucoseLacticaseibacillus casei 01
(paraprobiotic)
None[42]
↓ FPGLactobacillus acidophilus La-5 and
Bifidobacterium animalis subsp
lactis BB-12
T2D[43]
Lactobacillus acidophilus La-5 and
Bifidobacterium animalis subsp
lactis BB-12
Metabolic syndrome[44]
Bifidobacterium longum APC1472Healthy overweight/obese[45]
Lactobacillus plantarum OLL2712Pre-diabetic[46]
Ecologic® Barrier
(multi-strain probiotics)
T2D[47]
Lactobacillus casei 01T2D[48]
Lactobacillus caseiT2D[49]
↓ HbA1cLactobacillus acidophilus La-5 and
Bifidobacterium animalis subsp
lactis BB-12
T2D[50]
Lactobacillus reuteri ADR-1T2D[51]
“Symbiter” (multi-strain)T2D[52]
Lactobacillus acidophilus La-5 and
Bifidobacterium animalis subsp
lactis BB-12
T2D[43]
Lactobacillus acidophilus La-5 and
Bifidobacterium animalis subsp
lactis BB-12
T2D[53]
HOMA-IR improvement
(reduction)
Lactobacillus plantarum OLL2712Pre-diabetic[46]
“Symbiter” (multi-strain)T2D[52]
Ecologic® Barrier
(multi-strain probiotics)
T2D[47]
Lactobacillus casei 01T2D[48]
Lactobacillus caseiT2D[49]
Lactobacillus acidophilus La-5 and
Bifidobacterium animalis subsp
lactis BB-12
Metabolic syndrome[44]
↓ FructosamineLactobacillus acidophilus La-5 and
Bifidobacterium animalis subsp
lactis BB-12
T2D[50]
↓ Fasting insulin levelsEcologic® Barrier
(multi-strain probiotics)
T2D[47]
Lactobacillus caseiT2D[49]
Lactobacillus casei 01T2D[48]
Lactobacillus acidophilus La-5 and
Bifidobacterium animalis subsp
lactis BB-12
Metabolic syndrome[44]
Insulin sensitivity
improvement (↑ ISI, QUICKI test
or others)
Lactobacillus reuteri DSM 17938T2D[54]
Ecologic® Barrier
(multi-strain probiotics)
T2D[47]
Lactobacillus acidophilus NCFMT2D[55]
Lactobacillus plantarum OLL2712Pre-diabetic[46]
Lactobacillus acidophilus La-5 and
Bifidobacterium animalis subsp
lactis BB-12
Metabolic syndrome[44]
↓ GhrelinBifidobacterium longum APC1472Healthy overweight/obese[45]
↓ CortisolBifidobacterium longum APC1472Healthy overweight/obese[45]
↓ GlycoalbuminLactobacillus plantarum OLL2712Pre-diabetic[46]
↓ Fetuin-ALactobacillus casei 01T2D[48]
Lactobacillus caseiT2D[49]
↑ SIRTsLactobacillus casei 01T2D[48]
Lactobacillus caseiT2D[49]
↓ IL-6Lactobacillus plantarum OLL2712Pre-diabetic[46]
Ecologic® Barrier
(multi-strain probiotics)
T2D[47]
↓ TNF-α“Symbiter” (multi-strain)T2D[52]
Lactobacillus plantarum OLL2712Pre-diabetic[46]
Lactobacillus acidophilus La-5
and Bifidobacterium animalis subsp lactis BB-12
T2D[50]
Ecologic® Barrier
(multi-strain probiotics)
T2D[47]
↓ IL-1β“Symbiter” (multi-strain)T2D[52]
Lactobacillus plantarum OLL2712Pre-diabetic[46]
Lactobacillus reuteri ADR-3T2D[51]
↓ MCP-1Lactobacillus plantarum OLL2712Pre-diabetic[46]
T2D: type 2 diabetes; FPG: fasting plasma glucose; HbA1c: glycohemoglobin; HOMA-IR: homeostasis model assessment-estimated insulin resistance; ISI: insulin sensitivity index; HFD: high-fat diet; QUICKI: quantitative insulin sensitivity check index; OGTT: Oral Glucose Tolerance Test; SIRTs: sirtuins; IL-6: interleukin-6; IL-1β: interleukin 1 beta; TNF-α: tumour necrosis factor alpha; MCP-1: monocyte chemotactic protein-1; ↓: decrease; ↑: increase.
Table 2. Positive probiotic effects on some blood parameters in selected animal models studies.
Table 2. Positive probiotic effects on some blood parameters in selected animal models studies.
Altered Blood
Parameters
Type of ProbioticSpeciesPhysiological/Pathological Condition and/or DietReference
↓ Postprandial blood glucoseLactobacillus rhamnosus (YC, 7-1), Bifidobacterium adolescentis (N3, 7-2) and Bifidobacterium bifidum (M2)MouseHFD
STZ-induced diabetes
[77]
Lactobacillus plantarum CCFM0236MouseSTZ-induced diabetes[78]
↓ FPGLactobacillus casei and Bifidobacterium bifidum (alone or
combination of both)
ratSTZ-induced diabetes[79]
Lactobacillus plantarum TN627RatAlloxan-induced diabetes[80]
Bifidobacterium animalis 01RatHigh-fat chow diet
STZ-induced diabetes
[81]
Lactobacillus paracasei HII01RatHFD
STZ-induced diabetes
[82]
Lactobacillus rhamnosus (YC, 7-1), Bifidobacterium adolescentis (N3) and Bifidobacterium bifidum (M2)MouseHFD
STZ-induced diabetes
[77]
Lactobacillus rhamnosus (BSL and R23)RatSTZ-induced diabetes[83]
Heat-inactivated Streptococcus
thermophilus
RatHFD
ZDF diabetes
[84]
Lactiplantibacillus plantarum IMC 510RatDiet-induced obesity[85]
Lactobacillus fermentum RS-2RatAlloxan-induced diabetes[86]
Bacillus licheniformis
Zhengchangsheng®
MouseHFD-induced obesity[87]
Weissella cibaria MG5285,
Limosilactobacillus reuteri MG5149, Lacticaseibacillus rhamnosus MG4502, Lactobacillus gasseri MG4524
MouseHFD-induced obesity[88]
ProbiogluTMRatSTZ-induced diabetes[89]
Bacillus toyonensis SAU-19MouseHFD
ZDF-induced diabetes
[90]
Lactobacillus casei LC89MouseSTZ-induced diabetes[91]
Lactibiane Tolérance®MouseHFD[92]
Lactobacillus salivarus AP-32
Lactobacillus reuteri GL-104
MouseDb/db obesity[93]
Lactobacillus plantarum CCFM0236MouseSTZ-induced diabetes[78]
Lactobacillus plantarum YJ7MouseHFD[94]
Bifidobacterium longum BR-108
(inactivated)
MouseObese diabetes[95]
L. rhamnosus L12, L. acidophilus, L. plantarum HM218749, B. animalis subsp. lactis LPL-RH, B. longum subsp. longum BAMA-B05/BAu-B1024)MouseGlucose
STZ-induced diabetes
[96]
L. rhamnosus L12, L. acidophilus, L. plantarum HM218749, B. animalis subsp. lactis LPL-RH, B. longum subsp. longum BAMA-B05/BAu-B1024)MouseGlucose water
-induced diabetes
[96]
Lactobacillus plantarum DSM 15313MouseHFD-induced obesity[97]
Lactobacillus plantarum OLL2712MouseHFD-induced obesity[98]
Lactobacillus casei Shirota YIT 9029MouseHFD-induced obesity[99]
14 composite probioticsMouseDb/db diabetes[100]
14 composite probioticsMouseDb/db diabetes[101]
Bifidobacterium longumMouseHFD-induced obesity[102]
Lactobacillus plantarum HAC01MouseHFD
STZ-induced diabetes
[103]
Lactobacillus paracasei NL41RatHFD
STZ-induced diabetes
[104]
Akkermansia municiphila
and its extracellular vesicles
MouseHFD[105]
VSL#3 composite probioticsMouseHFD-induced obesity[106]
Lactobacillus paracasei DTA81MouseHFD[107]
Lactobacillus acidophilus SJLH001MouseHFD[108]
Lactobacillus casei ZhangRatHyperinsulinemia[109]
Lactobacillus acidophilus KLDS1.1003 and KLDS1.0901MouseHFD
STZ-induced diabetes
[110]
↓ HbA1cLactobacillus casei and Bifidobacterium bifidum (alone or combination of both)RatSTZ-induced diabetes[79]
Lactobacillus reuteri ADR-1RatHigh-fructose diet[51]
Bifidobacterium animalis 01RatHigh-fat chow diet
STZ-induced diabetes
[81]
Heat-inactivated Streptococcus
thermophilus
RatHFD
ZDF diabetes
[84]
Lactobacillus plantarum CCFM0236MouseSTZ-induced diabetes[78]
14 composite probioticsMouseDb/db diabetes[100]
14 composite probioticsMouseDb/db diabetes[101]
Lactobacillus plantarum HAC01MouseHFD
STZ-induced diabetes
[103]
Lactobacillus paracasei NL41RatHFD
STZ-induced diabetes
[104]
Lactobacillus acidophilus KLDS1.1003 and KLDS1.0901MouseHFD
STZ-induced diabetes
[110]
SLAB51 composite probioticsMouseAD[111]
HOMA-IR improvement (reduction)Bifidobacterium animalis 01RatHigh-fat chow diet
STZ-induced diabetes
[81]
Lactobacillus paracasei HII01RatHFD
STZ-induced diabetes
[82]
Lactobacillus rhamnosus (YC), Bifidobacterium adolescentis (N3) and Bifidobacterium bifidum (M2)MouseHFD
STZ-induced diabetes
[77]
Heat-inactivated Streptococcus
thermophilus
RatHFD
ZDF diabetes
[84]
Bacillus licheniformis
Zhengchangsheng®
MouseHFD-induced obesity[87]
ProbiogluTMRatSTZ-induced diabetes[89]
Bacillus toyonensis SAU-19MouseHFD
ZDF-induced diabetes
[90]
Lactobacillus casei LC89MouseSTZ-induced diabetes[91]
Lactibiane Tolérance®MouseHFD[92]
Lactobacillus plantarum CCFM0236MouseSTZ-induced diabetes[78]
Lactobacillus plantarum YJ7MouseHFD[94]
Lactobacillus plantarum DSM 15313MouseHFD-induced obesity[97]
14 composite probioticsMouseDb/db diabetes[100]
14 composite probioticsMouseDb/db diabetes[101]
Lactobacillus plantarum HAC01MouseHFD
STZ-induced diabetes
[103]
Lactobacillus paracasei NL41RatHFD
STZ-induced diabetes
[104]
VSL#3 composite probioticsMouseHFD-induced obesity[106]
Lactobacillus acidophilus KLDS1.0901MouseHFD
STZ-induced diabetes
[110]
HOMA-B reductionRomboutsia ilealis DSM 25109MouseWestern diet-induced diabetes[112]
↓ fasting insulin levelsLactobacillus paracasei HII01RatHFD
STZ-induced diabetes
[82]
Heat-inactivated Streptococcus
thermophilus
RatHFD
ZDF diabetes
[84]
Bacillus licheniformis
Zhengchangsheng®
MouseHFD-induced obesity[87]
ProbiogluTMRatSTZ-induced diabetes[89]
Lactobacillus casei LC89MouseSTZ-induced diabetes[91]
Romboutsia ilealis DSM 25109MouseWestern diet-induced diabetes[112]
Lactibiane Tolérance®MouseHFD[92]
Lactobacillus plantarum CCFM0236MouseSTZ-induced diabetes[78]
Lactobacillus acidophilus KLDS1.0901MouseHFD
STZ-induced diabetes
[110]
↑ insulin levelsLactobacillus casei and Bifidobacterium bifidum (alone or combination of both)RatSTZ-induced diabetes[79]
Lactobacillus rhamnosus (BSL and R23)RatSTZ-induced diabetes[83]
14 composite probioticsMouseDb/db diabetes[100]
Insulin sensitivity improvement (↑ ISI, QUICKI test or others)Lactobacillus rhmanosus (GG, YC), Bifidobacterium adolescentis (7-2) and Bifidobacterium bifidum (35)MouseHFD
STZ-induced diabetes
[77]
Lactobacillus rhamnosus (BSL and R23)RatSTZ-induced diabetes[83]
Bacillus amyloliquefaciens SC06MouseHFD-induced obesity[113]
Lactobacillus plantarum CCFM0236MouseSTZ-induced diabetes[78]
Lactobacillus plantarum DSM 15313MouseHFD-induced obesity[97]
Lactobacillus acidophilus KLDS1.0901MouseHFD
STZ-induced diabetes
[110]
Glucose tolerance improvement
(OGTT)
Bifidobacterium longum APC1472MouseHFD[45]
Lactobacillus casei and Bifidobacterium bifidum (alone or combination of both)RatSTZ-induced diabetes[79]
Lactobacillus rhmanosus, Bifidobacterium adolescentis and Bifidobacterium bifidum (all together 9 strains)MouseHFD
STZ-induced diabetes
[77]
Lactobacillus paracasei HII01RatHFD
STZ-induced diabetes
[82]
Bifidobacterium animalis 01RatHigh-fat chow diet
STZ-induced diabetes
[81]
Lactobacillus rhamnosus (BSL and R23)RatSTZ-induced diabetes[83]
Heat-inactivated Streptococcus thermophilusRatHFD
ZDF diabetes
[84]
Weissella cibaria MG5285,
Limosilactobacillus reuteri MG5149, Lacticaseibacillus rhamnosus MG4502, Lactobacillus gasseri MG4524
MouseHFD-induced obesity[88]
Romboutsia ilealis DSM 25109MouseWestern diet-induced diabetes[112]
ProbiogluTMRatSTZ-induced diabetes[89]
Lactobacillus salivarus AP-32
Lactobacillus reuteri GL-104
MouseDb/db obesity[93]
Bifidobacterium longum BR-108 (inactivated)MouseObese diabetes[95]
L. rhamnosus L12, L. acidophilus, L. plantarum HM218749, B. animalis subsp. lactis LPL-RH, B. longum subsp. longum BAMA-B05/BAu-B1024)MouseGlucose
STZ-induced diabetes
[96]
L. rhamnosus L12, L. acidophilus, L. plantarum HM218749, B. animalis subsp. lactis LPL-RH, B. longum subsp. longum BAMA-B05/BAu-B1024)MouseGlucose water-
Induced diabetes
[96]
Lactobacillus rhamnosus GGMouseDb/db obesity[114]
Bifidobacterium longumMouseHFD-induced obesity[102]
Lactobacillus plantarum HAC01MouseHFD
STZ-induced diabetes
[103]
Bifidobacterium longum OLP-01MouseHFD-induced obesity[115]
Limosilactobacillus fermentum MG4231 and MG4244MouseHFD-induced obesity[116]
Lactobacillus paracasei NL41RatHFD
STZ-induced diabetes
[104]
VSL#3 composite probioticsMouseHFD-induced obesity[106]
Lactobacillus acidophilus SJLH001MouseHFD[108]
Lactobacillus casei ZhangRatHyperinsulinemia[109]
↑ osteocalcinLactobacillus casei ZhangRatHyperinsulinemia[109]
↓ IL-6Heat-inactivated Streptococcus
thermophilus
RatHFD
ZDF-induced diabetes
[84]
ProbiogluTMRatSTZ-induced diabetes[89]
Lactobacillus plantarum YJ7MouseHFD-induced obesity[94]
Bacillus amyloliquefaciens SC06MouseHFD-induced obesity[113]
Lactobacillus casei LC89MouseSTZ-induced diabetes[91]
↓ TNF-αLactobacillus plantarum YJ7MouseHFD-induced obesity[94]
Bacillus amyloliquefaciens SC06MouseHFD-induced obesity[113]
Heat-inactivated Streptococcus
thermophilus
RatHFD
ZDF-induced diabetes
[84]
ProbiogluTMRatSTZ-induced diabetes[89]
Lactobacillus casei LC89MouseSTZ-induced diabetes[91]
SLAB51 composite probioticsMouseAD[117]
Bifidobacterium animalis 01RatHigh-fat chow diet
STZ-induced diabetes
[81]
Lactobacillus plantarum CCFM0236MouseHFD
STZ-induced diabetes
[78]
↓ IL-1βLactobacillus casei LC89MouseSTZ-induced diabetes[91]
Lactobacillus plantarum YJ7MouseHFD-induced obesity[94]
Heat-inactivated Streptococcus
thermophilus
RatHFD
ZDF-induced diabetes
[84]
ProbiogluTMRatSTZ-induced diabetes[89]
SLAB51 composite probioticsMouseAD[117]
↑ IL-10Heat-inactivated Streptococcus
thermophilus
RatHFD
ZDF-induced diabetes
[84]
Lactobacillus casei LC89MouseSTZ-induced diabetes[91]
Bifidobacterium animalis 01RatHigh-fat chow diet
STZ-induced diabetes
[81]
Lactobacillus plantarum CCFM0236MouseHFD
STZ-induced diabetes
[78]
T2D: type 2 diabetes; FPG: fasting plasma glucose; HbA1c: glycohemoglobin; HOMA-IR: homeostasis model assessment-estimated insulin resistance; HOMA-B: index reflecting pancreatic beta-cell function; ISI: insulin sensitivity index; QUICKI: quantitative insulin sensitivity check index; OGTT: oral glucose tolerance test; IL-6: interleukin-6; IL-10: interleukin 10; IL-1β: interleukin 1 beta; TNF-α: tumour necrosis factor alpha; ZDF: Zucker diabetic fatty; HFD: high-fat diet; STZ: streptozotocin; Db/db: genetically mutated model for type 2 diabetes with improper leptin receptor function; AD: Alzheimer’s disease; ↓: decrease; ↑: increase.
Table 3. Effect of probiotics on intestinal microbiota and glucose metabolism in in vivo animal model studies.
Table 3. Effect of probiotics on intestinal microbiota and glucose metabolism in in vivo animal model studies.
Animal Model/
Pathology
Administered
Probiotic
Altered Intestinal MicrobiotaEffect on
Glucose
Metabolism
Reference
Genera or Family
IncreaseDecrease
T2D mouseLactobacillus casei LC89Alloprevotella (B), Bacteroides (B), Parabacteroides (B),
Ruminococcus (F)
Lachnospiraceae
_NK4A136_group (F), Odoribacter (B)
Mucispirillum (D)
Glucose tolerance
improvement
[91]
HFD-
induced obese mouse
Lactobacillus
plantarum HT121
Akkermansia (V), Allobaculum (F), Lactobacillus (F), Prevotella (B),
Sutterella (P) and S24-7
family (B)
Anaerostipes (F)
Anaerotruncus (F)
Bilophila (P)
Candidatus_Arthromitus (F) Coprococcus (F), Dorea (F),
Mucispirillum (D),
Oscillospira (F),
Ruminococcus (F),
Streptococcus (F) and
families of
Peptococcaceae (F), Ruminococcaceae (F)
Glucose tolerance
improvement; blood
glucose decrease
[197]
HFD-
induced obese mouse
Lactobacillus
plantarum Q180
Rikenellaceae (B),
Ruminococcaceae (F),
Lachnospiraceae (F)
glucose tolerance
improvement
[142]
T2D mouseLactobacillus casei CCFM419Allobaculum (F),
Bacteroides (B)
Alleviation of type 2
diabetes
symptoms
(insulin
resistance and hyperglycaemia
amelioration)
[198]
T2D mouseLactobacillus
plantarum HAC01
Akkermansiaceae (V)Dusulfovibrionaceae (P)Reduction in glucose-mediated insulin
secretion
(SCFA
increase)
[103]
T2D mouse (db/db)14 composite probioticsBifidobacterium (A),
Lactobacillus (F),
Clostridium leptum (F),
Roseburia (F),
Prevotella (B),
Enterococcus faecium (F), Escherichia coli (P), Bacteroides
thetaiotaomicron (B)
Improvement in glucose
absorption
[101]
T2D ratLactobacillus G15 and Q14Clostridium leptum (F),
Bacteroides (B), Prevotella (B)
Bifidobacterium (A),
Lactobacillus (F)
Improvement in blood glucose and insulin
disorders
(glucose
tolerance)
[199]
T2D mouse*^Bifidobacterium
adolescentis N3,
*^Bifidobacterium
bifidum M2,
^Bifidobacterium
adolescentis 7-2,
°Lactobacillus
rhamnosus YC,
Lactobacillus
rhamnosus 7-1
* Bacteroidales S24-7 (B), ^Parabacteroides (B),
°Mucispirillum (D),
Coprococcus (F),
Streptococcus (F)
Glucose
metabolism and insulin
resistance
improvement
(reduction in blood glucose levels and
insulin
resistance,
regulation of SCFAs levels)
[77]
T2D mouseLactobacillus
acidophilus KLDS1.003 and KLDS1.0901
Blautia (F), Roseburia (F),
Anaerotruncus (F)
Desulfovibrio (T),
Alistipes (B)
Bacteroides (B)
Glucose and lipid metabolism-
related
signalling route
improvement of intestinal
microbiota
[110]
T2D ratStreptococcus
thermophilus
Ruminococcaceae (F),
Veillonella (F), Coprococcus (F), Barnesiella (B)
Moderation of insulin
resistance
(HOMA-IR, HbA1c improvement)
[84]
T2D
monkey
Lactobacillus
plantarum
- pMG36e-GLP-1
Alistipes (B)Prevotella (B)Blood glucose
reduction
(increase in SCFAs)
[200]
GDM
pregnant rat
Lactobacillus
rhamnosus LGG and
Bifidobacterium animalis subsp. lactis Bb12
Lachnospiraceae (F),
Dubosiella (F)
Muribaculaceae (F),
Ruminococcacea_
UCG-005 (F)
Carbohydrate
metabolism and
membrane transport
pathway
inhibition
[201]
T2D: type 2 diabetes; HFD: high-fat diet; db/db: db/db mouse has a mutation of the diabetes (db) gene encoding for the ObR; GDM: gestational diabetes mellitus; F: Firmicutes (Bacillota); B: Bacteroidetes (Bacteroidota); V: Verrucomicrobia (Verrucomicrobiota); D: Deferribacteres (Deferribacterota); P: Proteobacteria (Pseudomonadota); A: Actinobacteria (Actinomycetota); T: Thermodesulfobacteriota; HOMA-IR: homeostasis model assessment-estimated insulin resistance; HbA1c: glycohemoglobin; SCFA: short-chain fatty acids; *, ^, °, ’: each probiotic affects different types of bacteria according to the same symbol.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pintarič, M.; Langerholc, T. Probiotic Mechanisms Affecting Glucose Homeostasis: A Scoping Review. Life 2022, 12, 1187. https://doi.org/10.3390/life12081187

AMA Style

Pintarič M, Langerholc T. Probiotic Mechanisms Affecting Glucose Homeostasis: A Scoping Review. Life. 2022; 12(8):1187. https://doi.org/10.3390/life12081187

Chicago/Turabian Style

Pintarič, Maša, and Tomaž Langerholc. 2022. "Probiotic Mechanisms Affecting Glucose Homeostasis: A Scoping Review" Life 12, no. 8: 1187. https://doi.org/10.3390/life12081187

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