Effect of Probiotics on Glucose Metabolism and Homeostasis: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

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. 

  • probiotic
  • mechanism
  • glucose
  • metabolism
  • homeostasis

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 [1]. Microbiota acts at different levels through complex mechanisms that are not fully understood [2]. 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 [3];
  • Direct, contact-dependent principle (binding to different surface molecules);
  • Indirect principle via secretory molecules (production of bioactive peptides and metabolites).

2. Effect of Probiotics on Blood Parameters

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. [12] 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 [18] 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 [13]. In another 12-week human clinical trial, probiotic did not alter HbA1c levels, fat levels or total serum bile acids [16]. Furthermore, Shellekens et al. [7] 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 [19], 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 [8]. 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 [4].
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 [20]. The effect of probiotics on blood parameters was also demonstrated in a clinical trial conducted in the Arab population [9], 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 [9]. Kobyliak et al. [14] 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 [11] and Lactobacillus casei [10] 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 [6].
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 [14]. 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 [21], while tumour necrosis factor alpha (TNF-α) seems to be mainly involved in insulin signalling and action [22] and in the regulation of glucose and lipid metabolism [23]. 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 [14]. Toshimitsu et al. [8] 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) [24]. The mechanism by which probiotics reduce the amount of this parameter is still unclear [25]. Five clinical studies [5][12][13][14][15] 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 [11][26].
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 [27], as it affects the activity of insulin receptor tyrosine kinase [28]. Sirtuins (SIRTs) are very important regulators of energy homeostasis and metabolism due to their deacetylation activity in variety of organs [29]. 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 [10][11]. 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 [30]. 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 [30][31][32][33][34][35][36][37][38][39]. 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 [30][31][32][34][37], lack of data on dietary habits and physical activity of study participants [31][33][36], diversity within a study [32][34][35][36][37], short duration of probiotic therapy, different therapeutic doses, use of different probiotic strains [30][31][33][35][37], lack of evidence on possible side-effects of probiotic use [32] and lack of testing of other metabolic factors affecting glycaemic control [33][34]. Last, but not least, Tao et al. [39] proposed that more clinical data and research on the mechanisms of probiotic are needed.

2.2. In Vivo Animal Models Studies

Many studies in animal models demonstrated positive effects of various probiotics on diverse blood parameters. 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 [40]. Similar antidiabetic effects in rats were obtained by administration of probiotic strains Lactobacillus plantarum TN627 [41], Bifidobacterium animalis 01 [42], Lactobacillus paracasei HII01 [43], 9 strains of Lactobacillus rhamnosus, Bifidobacterium adolescentis and Bifidobacterium bifidum [44], Lactobacillus rhamnosus BSL and R23 [45], heat-inactivated Streptococcus thermophilus [46], Lactiplantibacillus plantarum IMC 510 [47] and Lactobacillus fermentum RS-2 [48]. In the latter case, in addition to reduced glucose levels in fasting state, a low oxidative stress was observed [48].
Studies in mice and rats have shown, among other things, a positive effect of probiotic preparations on glucose balance [49][50][51][52][53][54][55][56][57][58][59][60][61][62] and reduced insulin resistance [49][50][52][53][54][55][57][61]. Andersson et al. [63], Sakai et al. [64], Park et al. [65], Naito et al. [66], Wang et al. [67][68], Machado et al. [69], Lee et al. [70], Hsu et al. [71], Kim et al. [72] and Zeng et al. [73] have shown that the probiotic addition of different species of the genus Lactobacillus or a combination of probiotics [67][68][72] improves glucose sensitivity (tolerance) [63][64][65][66][67][68][69][70][71][72] and reduces insulin resistance [63][64][65][66][67][68] 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 [66].
Effects on glucose homeostasis and cholesterol metabolism have also been demonstrated by Ashrafian et al. [74], Wang et al. [75], Kim et al. [72], Tarrah et al. [76] and Sun et al. [77]. 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 [77]. Yan et al. [78] 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 [49][54][57][61][79] and rats [46][52], 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 [46], Bifidobacterium animalis 01 [42] and Lactobacillus casei LC89 [54], Lactobacillus plantarum CCFM0236 [49] 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 [42] 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 [79]. 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 [80].
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 [81]. However, it has been proven to have a positive effect on improving glucose tolerance and regulating glucose metabolism [81][82][83]. 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 [84].

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 [62]. In in vitro studies [49][57][62][77][85], certain probiotics inhibited α-glucosidase activity and thereby decreasing the level of glucose ready for absorption and thereby glycaemic index of food.

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 [86].
Recent studies have revealed an important role of glucose homeostasis in some brain pathological conditions, such as Alzheimer’s disease (AD) [79]. 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 [80].

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 [87].
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) [88]. In fact, FGF plays an important role in the regulation of bile acid synthesis and its amount directly influences lipid and glucose metabolism [89].
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 [16].
In a study in rats the probiotic Lactobacillus casei Zhang [84] 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 [90]. In another study conducted in rats [91], the same probiotic influenced the reduction of bacteria with hydrolase activity, resulting in increased bile acid secretion.

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 [92][93] (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 [94].
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.
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 [95]. In the second study in mice, Lactobacillus sakei OC67 reduced the incidence of hyperglycaemia and obesity by lowering inflammation [96]. In both studies, as well as in three other studies [64][97][98] 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 [97], Lactobacillus crustorum MN047 and Lactobacillus rhamnosus LS-8 [98]. The later probiotic decreased the IL-6 expression in adipose tissue as well [98]. 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 [65].
Deng et al. [99] suggested that different genotypes of Akkermansia municiphila exert specific effects in repairing damage to adipose tissue caused by a high-fat diet in mice. That 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 [100] and Bacillus [61] 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 [101]. Increased adiponectin mRNA levels in fat cells (adipocytes) [100] and increased serum leptin levels [61] 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 [102]. However, in a study in mice [103], 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 [104]. 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 [103]. Indeed, Molina-Tijeras et al. [105] 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 [106].
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 [107]. In a study on rats [84], 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 [108], while GLUT4 is the major glucose transporter for the facilitated transfer of glucose from blood to skeletal fat tissue and muscle [109]. 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 [105]. In addition to PPAR-γ, some probiotics have also been shown to affect increased expression of PPAR-α. in animal models [74][110] and in one in vitro study [111]
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 [112][113]. 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 [74].
Lactobacillus plantarum Ln4 significantly reduced lipid deposition and induced glucose absorption in 3T3-L1 adipocytes in an in vitro study [114].

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 [115]. 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 [65][95].
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 [40]. 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 [103].
The protective effect of Lactobacillus plantarum was demonstrated in a study conducted on mice [106]. 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. [116] 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 [117] and rats [43][110]. Toejing et al. [43] 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.
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.

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 [118]. Studies on animal models have shown that single probiotics reduce oxidative stress in the pancreas [40][73][119], liver [53][56][73][120] and kidney [56][73], thus helping to increase the antioxidant activity and physiological function of the organ.

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 3 shows some common effects of probiotics in the intestine and the impact of their action on glucose metabolism and homeostasis.
Figure 3. 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.

This entry is adapted from the peer-reviewed paper 10.3390/life12081187

References

  1. 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.
  2. Plaza-Diaz, J.; Ruiz-Ojeda, F.J.; Gil-Campos, M.; Gil, A. Mechanisms of Action of Probiotics. Adv. Nutr. 2019, 10, S49–S66.
  3. Suez, J.; Zmora, N.; Segal, E.; Elinav, E. The pros, cons, and many unknowns of probiotics. Nat. Med. 2019, 25, 716–729.
  4. 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.
  5. 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.
  6. 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.
  7. 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.
  8. 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.
  9. 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.
  10. 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.
  11. 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.
  12. 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.
  13. 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.
  14. 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.
  15. 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.
  16. 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.
  17. 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.
  18. 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.
  19. 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.
  20. 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.
  21. Holmes, D. Physiologic role of IL-1β in glucose homeostasis. Nat. Rev. Endocrinol. 2017, 13, 128.
  22. 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.
  23. 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.
  24. 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.
  25. 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.
  26. 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.
  27. 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.
  28. 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.
  29. Turkmen, K.; Karagoz, A.; Kucuk, A. Sirtuins as novel players in the pathogenesis of diabetes mellitus. World J. Diabetes 2014, 5, 894.
  30. 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.
  31. 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.
  32. 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.
  33. 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.
  34. 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.
  35. 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.
  36. 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.
  37. Kasińska, M.A.; Drzewoski, J. Effectiveness of probiotics in type 2 diabetes: A meta-analysis. Pol. Arch. Med. Wewn. 2015, 125, 803–813.
  38. 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.
  39. 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.
  40. 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.
  41. 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.
  42. 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.
  43. 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.
  44. 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.
  45. 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.
  46. 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.
  47. 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.
  48. 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.
  49. 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.
  50. 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.
  51. 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.
  52. 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.
  53. 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.
  54. 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.
  55. 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.
  56. 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.
  57. 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.
  58. Ben Othman, M.; Sakamoto, K. Effect of inactivated Bifidobacterium longum intake on obese diabetes model mice (TSOD). Food Res. Int. 2020, 129, 108792.
  59. 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.
  60. 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.
  61. 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.
  62. 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.
  63. 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.
  64. 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.
  65. 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.
  66. 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.
  67. 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.
  68. 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.
  69. 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.
  70. 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.
  71. 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.
  72. 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.
  73. 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.
  74. 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.
  75. 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.
  76. 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.
  77. 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.
  78. 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.
  79. 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.
  80. 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.
  81. Komori, T. Functions of Osteocalcin in Bone, Pancreas, Testis, and Muscle. Int. J. Mol. Sci. 2020, 21, 7513.
  82. Zoch, M.L.; Clemens, T.L.; Riddle, R.C. New insights into the biology of osteocalcin. Bone 2016, 82, 42–49.
  83. 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.
  84. 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.
  85. 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.
  86. Stavropoulou, E.; Bezirtzoglou, E. Probiotics in Medicine: A Long Debate. Front. Immunol. 2020, 11, 2192.
  87. 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.
  88. 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.
  89. 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.
  90. 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.
  91. 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.
  92. 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.
  93. 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.
  94. Hotamisligil, G.S. Inflammation and metabolic disorders. Nature 2006, 444, 860–867.
  95. 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.
  96. 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.
  97. 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.
  98. 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.
  99. 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.
  100. 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.
  101. Fang, H.; Judd, R.L. Adiponectin Regulation and Function. Compr. Physiol. 2018, 8, 1031–1063.
  102. 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.
  103. 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.
  104. 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.
  105. 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.
  106. 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.
  107. 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.
  108. Olefsky, J.M.; Saltiel, A.R. PPAR gamma and the treatment of insulin resistance. Trends Endocrinol. Metab. 2000, 11, 362–368.
  109. Mueckler, M.; Thorens, B. The SLC2 (GLUT) family of membrane transporters. Mol. Aspects Med. 2013, 34, 121–138.
  110. 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.
  111. 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.
  112. 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.
  113. 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.
  114. 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.
  115. Flamment, M.; Hajduch, E.; Ferré, P.; Foufelle, F. New insights into ER stress-induced insulin resistance. Trends Endocrinol. Metab. 2012, 23, 381–390.
  116. 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.
  117. 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.
  118. Oberley, L.W. Free radicals and diabetes. Free Radic. Biol. Med. 1988, 5, 113–124.
  119. 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.
  120. 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.
More
This entry is offline, you can click here to edit this entry!
ScholarVision Creations