Open access peer-reviewed chapter

Probiotics in the Management of Diabetes

Written By

Akash Kumar, Jhilam Pramanik, Nandani Goyal, Bhupendra G. Prajapati and Dimple Chauhan

Submitted: 07 January 2023 Reviewed: 02 February 2023 Published: 21 February 2024

DOI: 10.5772/intechopen.110338

From the Edited Volume

Advances in Probiotics for Health and Nutrition

Edited by Vasudeo Zambare, Mohd Fadhil Md. Din, Puja Gupta and Bhupendra Gopalbhai Prajapati

Chapter metrics overview

48 Chapter Downloads

View Full Metrics

Abstract

Gut microflora that has been present in our bodies since infancy are known to influence health, metabolism, and disease. Diabetes is a developing epidemic, and treatment that cures diabetes has yet to be discovered. Probiotics are living bacteria that may colonize the human gastrointestinal system and help to maintain a healthy microbiome and help normalize disrupted metabolism in diabetic patients. Lipopolysaccharides, trimethylamine, and imidazole propionate seem to hinder insulin signaling, whereas secondary bile acids, short-chain fatty acids (SCFAs), and tryptophan metabolites seem to enhance it. This chapter emphasizes the relationship between gut microflora and impaired glucose metabolism. This chapter also covers the mechanisms through which probiotics alleviate diabetes by addressing the gut microflora from the perspectives of amino acid metabolism, intestinal permeability, immunological responses, oxidative stress, and SCFAs.

Keywords

  • diabetes
  • probiotics
  • gut microflora
  • short-chain fatty acids (SCFAs)
  • Lactobacillus
  • Bifidobacterium

1. Introduction

Diabetes mellitus (DM), often known as diabetes, is a metabolic syndrome caused by abnormalities in the body’s capacity to generate insulin and/or activate insulin, or both. Hyperglycemia and glucose intolerance are the symptoms of diabetes mellitus [1]. Hyperglycemia increases the complications in the microvascular system (neuropathy, retinopathy, and nephropathy) as well as in the macrovascular system (stroke, ischemic heart disease, and peripheral vascular disease). As a result, there is a marked increase in morbidity and a significant decline in the quality of life [2, 3, 4]. According to the World Health Organization (WHO), 420 million people worldwide have diabetes, and the prevalence was estimated to be 2.8% in 2000, rising to 4.8% by 2030. Over the last two decades, the disease has been more prevalent than expected [5]. DM is seen as a huge global health and economic burden in the aging population and is now the eighth biggest cause of mortality globally [6]. The number of diabetics in India alone is already over 40 million, and by 2030, the country will have the biggest diabetic population in the world with a population of over 90 million [7, 8].

Diabetes has three basic subtypes: type 1, type 2, and gestational diabetes [9, 10, 11]. About 10% of all cases of diabetes are type 1 diabetes (T1DM), which is characterized by the impairment of insulin-secreting B-cells and needs daily insulin therapy for survival [12, 13]. T1DM is becoming more common in the world as a result of ineffective preventative and treatment approaches. Therefore, a thorough understanding of T1DM’s pathophysiology is necessary. Environmental factors and genetic factors play a crucial role in the progression of T1DM [14, 15]. Most people with diabetes (90–95%) have type 2 diabetes (T2DM), which is defined by impaired lipid and glucose metabolism brought on by insufficient insulin production or by its insensitivity [1]. Although T2DM is mostly diagnosed in older persons, the frequency of the condition in youngsters has been shown to rise as a result of obesity and physical inactivity [16]. Smoking, hereditary factors, excessive calorie consumption, and sedentary lifestyle are the main risk factors for T2DM, with alteration in gut microbiota as one of the causes and associated comorbidities [17]. A common problem that affects roughly 2–5% of all pregnancies, gestational diabetes mellitus (GDM) is characterized by high glucose levels in the second and third trimesters of pregnancy. It may manifest as either type I or type II diabetes in persons who have an inherited tendency to have the disease [18]. In the future, T2DM is more likely to occur in women with GDM due to their increased risk of pregnancy problems and premature birth [19, 20].

As per epidemiological observations, one of the characteristics of diabetic patients is changes in the diversity of intestinal microflora. Additionally, there is increasing evidence that diabetes and intestinal microflora are closely related. The microflora, host cells, and nutrients make a complex ecosystem that creates up the human gut. The alimentary canal contains about 100 trillion bacteria, which together make up the intestinal flora [21]. The bacteria that make up the intestinal flora are numerous and diverse. Genus, family, order, and phylum classifications are used to taxonomically group these. In healthy adults, the six phyla Firmicutes, Proteobacteria, Bacteroidetes, Actinobacteria, Fusobacteria, and Verrucomicrobia make up most of the intestinal microflora [22]. Researchers have shown that gut microbiota in diabetics is less reliable than in healthy individuals. In a sick condition functionality of gut microbiota changes, a human metagenome-wide association study conducted in Europe and China found surprising connections between specific bacterial genes, gut microbes, and the digestive system in T2DM patients [23]. These individuals showed greater levels of Lactobacillus spp. than nondiabetics, and fasting glucose and glycated hemoglobin (HbA1c) levels are positively connected with these levels [24]. Clostridium spp. had a negative relationship with fasting blood sugar and plasma triglycerides [25]. According to one investigation, it has been found that the number of Prevotella and Faecalibacterium decreased in diabetic conditions and demonstrated that the microbiome impacts both T1DM and T2DM [26]. In the mucous layer, there is an increase in Akkermansia muciniphila after metformin therapy [27]. It has been hypothesized that type 1 diabetes (T1DM) and autoimmune diabetes may both develop due to inflammation [28]. Autoimmune diabetes has been related to the microbiota of the gut because of the common receptors in the inflamed pancreas and the gut [29].

Diabetes interventions include medication [30], nutritional care [31, 32], physical activity [32], or weight control [33, 34]. They might also involve education, coaching, or social support [35]. As stated above, diabetes affects the gut microbiome; therefore, probiotics can be employed as one of the nutritional interventions. These are live bacteria that are given in sufficient amounts and continue to remain in the gut bionetwork to have a beneficial impact on one’s health [36]. Lilly and Stillwell used the word “probiotics” to refer to “organisms and substances which contribute to intestinal microbial balance” [37]. Probiotics are “organisms and compounds that help to gut microbial equilibrium,” according to Parker [38]. The International Scientific Association for Probiotics and Prebiotics (ISAPP), which was supported by the Food and Agriculture Organization of the United Nations/World Health Organization (FAO/WHO, 2001), defined probiotics as “Live microorganisms which, when administered in adequate amounts, confer a health benefit on the host” [39]. Probiotics are defined by the World Health Organization (WHO) as “products or preparations containing live, designated microorganisms in appropriate quantities that give positive effects on the host by altering its gut microbiota” [40].

Probiotics play an important role in immune system development, immune system homeostasis, and epithelial cell differentiation and proliferation [41]. Probiotics are not a recent discovery but have been present in many of our traditional foods for a long time, including drinks, salty fish, yogurt, various types of cheese, and so forth. Before the invention of the microscope, people were able to prepare a variety of milk products with various flavors and structures [42]. This is the result of various microbial reactions brought on by various microbes [43]. We really had no idea how probiotic-containing foods were first used, especially for therapeutic purposes. It is possible that Ilya Ilyich Metchnikoff, who won the Nobel Prize in Medicine in 1908, was the first to notice the effects of what is now known as probiotics while working at the Pasteur Institute. He correlated the consumption of yogurt’s microorganisms with good health. He proposed in 1907 that the bacteria Lactobacillus bulgaricus and Streptococcus thermophilus, which are involved in yogurt fermentation, block the putrefactive-type fermentations of the intestinal flora. He linked the consumption of yogurt containing the Lactobacillus species to the longevity and good health of Bulgarian peasants, and he presented his findings to the public in a manner that was easily understood [44].

The ISAPP consensus panel explained the concept that some probiotic mechanisms may be expressed by most strains of a larger taxonomic group, which is an evolving idea regarding the strain specificity of probiotic effects [45]. Lactic acid bacteria (LAB) are a group of predominant gut-friendly bacteria found in the digestive tract [46] and suppress pathogens through their secretions [47]. For instance, the majority of Bifidobacterium and Lactobacillus species both produce organic acids like lactate and acetate. There are several potential advantages for the gastrointestinal system. The inhibition of harmful microbes and the cross-feeding of other advantageous gut microbes result in the production of butyrate, which plays a significant role in cultivating a healthier gut environment [48]. The types of microbes from the genera Lactobacillus, Bifidobacterium, and Saccharomyces that are most frequently used as probiotics include these. Other genera of probiotics include Escherichia, Propionibacterium, Streptococcus, and Bacillus. Probiotics are poised to be an important tool for influencing the gut ecosystem’s function to enhance the nutritional status and health [49, 50]. The mechanisms of action that researchers have identified in various probiotic strains against diabetes are shown in Figure 1. However, there are still a lot of gaps in our understanding of the mechanisms underlying health benefits.

Figure 1.

Potential mechanisms linking probiotics to diabetes.

Modification of the gut microbiota’s composition is one alleged probiotic effect that has been challenging to prove in healthy humans. Although it is widely believed that probiotics “support a healthy intestinal flora,” [51], probiotic organisms seldom survive for longer than a few weeks after consumption [52]. Alpha diversity, richness, and evenness of the fecal microbiota were examined in a systematic review of studies looking at the effects of probiotics [51].

Research is still finding links between the microbiota and diabetes, and these seem to involve a variety of metabolic and immune response processes, most of which are linked to more specific mechanisms. Future investigations into the relationship between variations in the gut microbiota balance and diabetes may result in new interventional studies. This review provides an overview of the role of probiotics in diabetes management.

Advertisement

2. Probiotic interventions to ameliorate T1DM

Probiotics may, thus, be useful in T1DM prevention and management. By altering the gut microbiota, certain probiotic strains exhibit a positive impact on host health by boosting the synthesis of advantageous metabolites [53]. Additionally, by activating free fatty acid receptor 2 (FFAR2) and free fatty acid receptor 3 (FFAR3), which are involved in the regulation of the immune system and the pathogenesis of autoimmune diseases like T1DM, the administration of probiotic strains may increase the production of SCFAs (such as butyrate) and thereby balance the intestinal cellular homeostasis [54]. Additionally, intestinal L-cells’ ability to produce glucagon-like peptide-1 (GLP-1) might be improved by the activation of FFAR2/3 by SCFAs. The hormone GLP-1 is known as the “incretin effect” because it promotes the release of insulin from pancreatic beta-cells, lowering blood sugar levels [55, 56]. These findings show how probiotics may prevent or manage T1DM by preserving or re-establishing the gut microbiota-immune axis’ equilibrium. Figure 2 shows an overview of potential mechanisms of probiotics against type 1 diabetes.

Figure 2.

Potential mechanisms of probiotics against type 1 diabetes.

2.1 Animal studies

Lactobacillus brevis strains protect mice (streptozotocin [STZ]-induced T1DM) and lower blood glucose levels via the action of gamma-aminobutyric acid (GABA) [57]. It has been shown that probiotic strains from the families Bifidobacteriaceae and Lactobacillaceae and the genus Streptococcus thermophilus reduce intestinal inflammation, alleviate T1DM, and maintain gut immunological homeostasis by blocking IL-1 expression in nonobese diabetic (NOD) mice [58]. The feeding of a Lactobacillus lactis strain has also been demonstrated to have preventative benefits against T1DM development in NOD mice by the activation of the production of anti-inflammatory cytokines. Interestingly, the combination of the L. lactis strain with modest dosages of anti-CD3 boosted the production of IL-10. The intervention also led to the formation of antigen-specific Foxp3+ Tregs, which preserves pancreatic islets [59, 60]. Bifidobacterium species change the cytokine secretion pattern in gut-associated lymphatic tissue (GALT) from a proinflammatory condition to an anti-inflammatory. Controlling the preservation of the variety of B-cells and lowering insulitis reduced the likelihood of developing an islet-specific autoimmune disease and offered protection against autoimmune T1DM [61]. Table 1 shows an overview of important studies demonstrating the effect of probiotics on an animal model with type 1 diabetes mellitus.

ProbioticsModel typeMechanism of actionMajor findingsReferences
Oral probiotics VSL#3Nonobese diabetic mouse modelProducing more IL-33, indoleamine 2,3-dioxygenase (IDO)
Decreasing the production of inflammatory cytokines
Encouraging differentiation of CD103+
Lowering Teff/Treg cell ratios in MLNs, PLNs, and gut mucosa
Alteration of the microbial ecology in the gut
Altering the pathophysiology of T1DM
[58]
Streptococcus salivarius, Lactobacilli, and BifibobacteriaNonobese diabetic mouse modelSlowing down the rate of cellular disruption
Pancreatic pseudocysts, the pancreas, and the spleen produce more IL-10.
Preventing autoimmune diabetes[61]
Probiotic fermented milk with 1% of Lactobacillus speciesStreptozotocin-induced albino Wistar rat modelA substantial reduction in the liver’s ability to express genes involved in gluconeogenesis
Significant reductions in IL-6 and TNF- levels in the serum;
Declines in blood sugar levels, HbA1c, and blood lipid profiles.
Raising insulin levels while significantly lowering blood sugar levels
Enhancement of glucose metabolism
Reduction of oxidative stress, inflammation, and hepatic gluconeogenesis
[62]
Lactobacillus brevisStreptozotocin-induced diabetic Mouse ModelHigh ability to produce GABA because of the gad gene
Significant reduction in plasma insulin levels or blood glucose levels
Preventing T1DM from developing in mice[57]
Bifidobacterium spp.Streptozotocin-induced diabetic Mouse ModelA dramatic drop in levels of blood sugar
Enhancing insulin receptor substrate, insulin receptor substrate 1, and expression of insulin receptor β
Reducing the expression of IL-6 and macrophage chemoattractant protein-1
Manage diabetes[63]
Lactobacillus reuteriStreptozotocin-induced diabetic Mouse ModelInhibition of osteoblast TNF-signaling results in the development of anti-inflammatory propertiesThe administration of probiotics may help T1DM patients’ bones[64]
Lactobacillus kefiranofaciens and Lactobacillus kefiriStreptozotocin-induced diabetic Mouse ModelPancreatic IL-10 level dramatically increased
More IL-10 prevents the production of TNF-α and TH1 and other pro-inflammatory cytokines
Capacity to induce GLP-1 release[65]
Lactobacillus johnsonii N6.2T1DM biobreeding diabetes-prone rats modelModifications to the gut’s natural microbiome
Induced oxidative stress response and alterations in host mucosal protein
Lowering intestinal mucosal oxidative response protein
Reducing cytokines that are proinflammatory
Increasing expression of claudin and other tight junction proteins
Delaying or preventing the onset of T1DM.[66]

Table 1.

Overview of important studies demonstrating the effect of probiotics on an animal model with type 1 diabetes mellitus.

Biobreeding diabetes resistant (BBDR) rats are more likely to develop T1DM when exposed to Kilham rat virus (KRV) infectious disease [67]. Similar results have also been seen in LEW.1WR1 rats that have had viral infections; these animals acquire autoimmune T1DM as a result of the infection of their cells. It has been shown experimentally that the oral administration of the Lactobacillus johnsonii strain develops resistance to the onset of T1DM in BBDR rats [67]. Consequently, L. johnsonii was linked to TH17 lymphatic cell predilection inside the mesenteric lymph nodes (MLNs) and might lower the incidence of T1DM in the bio-breeding diabetes-prone (BBDP) rat model. Increasing the level of the intestinal tight-junction protein claudin, L. johnsonii, also reduced the likelihood of developing T1DM [66]. In another experiment, probiotic-fermented milk was fed to diabetic rats induced by STZ. Consuming probiotic-fermented milk also reduced oxidative stress, inflammation, blood sugar levels, and the rate of gluconeogenesis [62]. Another investigation employing probiotic strain Lactobacillus plantarum in diabetic rats concluded that taking probiotics significantly decreased the serum α-amylase’s activity, favoring the glycemic index mechanism by limiting the digestion and hydrolysis of carbohydrates [68].

2.2 Human studies

Early exposure to probiotic supplements may reduce the incidence of islet-cell autoimmunity in relation to the increased risk of T1DM [69, 70, 71]. Additionally, probiotic usage has been linked to improved glucose control, increased GLP-1 production, and decreased TLR4 signaling in T1DM adults [72, 73, 74]. T1DM occurrences have reduced as a result of these modifications. Children with T1DM may benefit from taking Lactobacillus rhamnosus and Bifidobacterium lactis at a dosage of 109 colony-forming units (CFUs) once a day for six months regulates gut microbiota disturbances. Results indicated the modification of immune cells in a positive way and maintaining the quantity and proliferation of pancreatic β-cells [75]. Additionally, it has been suggested that adult human subjects consuming Lactobacillus johnsonii N6.2 (108 CFUs) in one capsule per day for eight weeks can control the natural killer cells and infiltration of monocytes. These modifications may help to prevent the development of T1DM. Furthermore, probiotic therapy has been linked to an increase in TH17 and TH1/TH17 cells. However, the probiotics-treated group showed a substantial rise in IgA concentration as compared to the placebo group [71]. Table 2 shows an overview of important studies demonstrating the effect of probiotics on human subjects with type 1 diabetes mellitus.

ProbioticsModel typeMechanism of actionMajor findingsReferences
Lactobacillus johnsoniiAdult humansIncreasing tryptophan levels in the serum
Lowering the plasma kynurenine to tryptophan ([Kyn]/[Trp]) ratio
Delaying or lessening the memory of CD8+ T-cell apoptosis
Lowering the risk of developing T1DM.[71]
Bifidobacterium lactis and Lactobacillus rhamnosusChildren (age range of 8–17)Enhancing the barrier property of the gastrointestinal mucosa
Decreasing autoimmunity risk
Altering the local and systemic immunological responses
Retaining the function of the—β-cell while inhibiting the proliferation of infections[75]
Probiotics and vitamin DChildren (age range of 4 to 10 years)Reducing the risk of islet autoimmunityEarly probiotic supplementation may reduce the risk of islet autoimmunity[70]
ProbioticsAdult humanDecreasing waist-to-hip ratio, body mass index, and obesity
Controlling triglyceride levels, HDL cholesterol, and blood pressure
Strongly linked to improve glycemic management
Beneficial impact on a range of variables connected to diabetes problems[76]

Table 2.

Overview of important studies demonstrating the effect of probiotics on human subjects with type 1 diabetes mellitus.

By using probiotics products, T1DM adult patients might improve their glycemic control and manage symptoms associated with metabolic syndromes, such as hypertension, elevated level of triglyceride, and decreased HDL levels. These findings together imply that probiotic intake may lower the likelihood of T1DM progression. Other research on young children with a genetic risk of T1DM consumed a probiotic strain during the first two years of life and the risk of the onset of islet autoimmunity and progression of T1DM was increased [77]. This suggests that all probiotic strains do not have the same effects, although the cause of the results from these studies is still unknown.

Advertisement

3. Probiotic interventions to ameliorate T2DM

Currently, probiotics from the former genera of Lactobacillus and Bifidobacterium are the major focus of most therapeutic investigations. However, specific bacterial strains are linked to the amelioration of disorders linked with inflammation. It is anticipated that some of the newly identified strains will become probiotics in the future [78]. Figure 3 shows the overview of potential mechanisms of probiotics against type 2 diabetes. Numerous studies have utilized probiotics in T2DM patients to manage or treat the disease, but the number of studies is still less. When analyzing these studies, it is necessary to use caution because it is well known that the effectiveness of probiotics depends on various factors such as:

  • The strain of microorganism (single or multistrain)

  • Pathophysiology of the disease

  • Subject of study

  • Type and quantity of dosage

  • Time period of intervention.

Figure 3.

Potential mechanisms of probiotics against type 2 diabetes.

3.1 Animal model study

Probiotics have been proven in studies to reduce insulin resistance (IR) in diabetic animal models (see Table 3). Probiotics like Lactobacillus spp. and Bifidobacterium spp. have been widely studied in diabetic animal models for their biological effects on glucose intolerance and insulin resistance (IR). Lactobacillus plantarum was given to the rats (which consume high-fat-diet [HFD]- and STZ-induced diabetes). It has been observed that L. plantarum reduces pancreatic beta-cell dysfunction, systemic inflammation, and insulin resistance [79]. L. plantarum reduced the weight and relieved IR in mice fed on the HFD [80]. In mice with diabetes generated by HFD, therapy with Lactobacillus fermentum has been demonstrated to reduce IR and stop the progression of diabetes [81]. The injection of Lactobacillus paracasei TD062 increased the insulin signaling pathway and improved glucose homeostasis, delaying the onset of T2DM [82]. In STZ-induced diabetic rats, a multiprobiotic formula, including Lactobacillus reuteri, Lactobacillus crispatus, and Bacillus subtilis, was studied. This research revealed that the daily consumption of probiotics may reduce glucose intolerance and increase insulin production [83]. By lowering fasting blood glucose (FBG), the oral glucose tolerance test (OGTT), and the HbA1c indices and increasing GLP-1 secretion, a composite probiotic made up of 10 Lactobacillus strains and four yeast strains were reported to improve T2DM in db/db mice [89]. Nano-selenium-enriched Bifidobacterium longum reduced the renal complication of T2DM in STZ-induced diabetes rats [84]. B. longum DD98 reduced the fasting blood glucose and HbA1c in HFD- and STZ-induced diabetic mice [90]. In diabetic rats caused by HFD and STZ, Bifidobacterium animalis administration increased oral glucose tolerance test and homeostatic model assessment for insulin resistance (HOMA-IR) indices and decreased proinflammatory cytokines [91].

ProbioticsModel usedMechanismOutcomesReferences
Lactobacillus plantarumHFD- and STZ-induced T2DM miceReducing systemic inflammation and insulin resistanceAmeliorate T2DM[79]
L. plantarum Ln4Mice fed on HFDModifications in hepatic gene expression that control lipid and glucose metabolism (reduced CD36 and increased mRNA levels for IRS2, Akt2, AMPK, and LPL)Reducing biomarkers for T2DM and obesity caused by diet[80]
L. plantarum and Lactobacillus fermentumMice fed with HFDPreventing the onset of insulin resistance and diabetes.Seem to be effective against T2DM[81]
Lactobacillus paracaseiHFD and STZ-induced T2DM miceControlling the levels of hepatic glycogen, lipid metabolism, glucose tolerance, postprandial blood glucose (PBG), and fasting blood glucose (FBG). Additionally, the antioxidant capability was enhancedPreventing the development of T2DM[82]
Lactobacillus reuteri, Lactobacillus crispatus, and Bacillus subtilisSTZ-induced diabetesAfter the intervention period, Glut-4 and PPAR-γ gene expression improved
Considerable elevation in insulin levels and a large reduction in plasma glucose and HbA1c values
Probiotics may help to manage diabetes and its complications if taken regularly[83]
Nano-selenium-enriched B. longumStreptozotocin-induced diabetesThe expression of insulin signaling pathway-related proteins was upregulated in the Nano-Se-B longum-treated groupsPreventive effect of Nano-Se-B longum on the onset of diabetes and renal damage[84]
Lactobacillus plantarumMice fed with HFDSignificantly reduced the mRNA expression of interleukin-1β in adipose tissue and serum levels of nonesterified fatty acids in miceSignificant reduction of blood glucose levels[85]
Lactobacillus caseiHFD- and STZ-induced T2DM miceDecreased levels of the inflammatory markers, tumor necrosis factor-α and interleukin-6 and increased intestinal glucagon-like peptide-1 (GLP-1) levels, which are associated with the production of short-chain fatty acids (SCFAs)Modifying the gut microbiota, increasing the production of SCFAs, and ameliorating type 2 diabetes[86]
Lactobacillus paracaseiHFD- and STZ-induced T2DM miceReducing the level of oxidative stress and insulin resistance, while also safeguarding beta-cell function and inhibiting the expansion of alpha-cellIndicating that the pancreatic islets as the key target tissues for the probiotic strain’s ameliorative action against T2DM[87]
Clostridium butyricumHFD- and STZ-induced T2DM miceIncreased insulin signaling molecules, and peroxisome proliferator-activated receptor (PPAR), as well as altered intestinal flora diversityTreating and preventing metabolic impairment caused by T2DM[88]

Table 3.

Overview of important studies demonstrating the effect of probiotics on an animal model with type 2 diabetes mellitus.

3.2 Human studies

Sabico et al. examined the effects of consuming 1010 CFU/day of a multistrain probiotic regarding metabolic endotoxemia levels and cardiometabolic parameters in adult patients recently diagnosed with T2DM. It has been found that the waist-hip ratio decreased across groups, while HOMA-IR was increased. The fasting blood glucose (FBG) level is less in the probiotic group when compared with the control group, while there are no substantial changes in the endotoxin levels [92]. In further research, the effect of the same probiotic mixture was examined for six months while using the same dosage and criteria as the earlier study. Again, a clinically substantial change in the HOMA-IR was noted, and the probiotic group’s insulin levels showed a borderline significant improvement [93]. When the flow of lipopolysaccharides (LPS) is decreased, it is anticipated that low-grade inflammation would decrease and insulin signaling will improve. Karczewski et al. assessed the effects of the probiotic Lactobacillus plantarum. The probiotic was injected directly into the duodenum of a group of people and followed by a tissue biopsy after 6 hours. According to the authors’ observations, zonula occludens-1 and occludin are translocated more often near tight junctions [94]. Similar results for various strains of the Lactobacillus genus were obtained in cell cultures [95]. In a nine-month double-blinded, randomized, placebo-controlled research, Hsieh et al. found that T2DM patients who consume capsules containing the probiotic Lactobacillus reuteri ADR-1 had lowered cholesterol and HbA1C level in their blood. The reduction in HbA1C was maintained even after three months of follow-up without probiotic treatment [96]. The effects of ingesting Lactobacillus reuteri for 12 weeks at various doses (low dose: 108 CFU/day vs. high dose: 1010 CFU/day) were examined by Mobini et al.; however, they were unable to detect a reduction in HbA1C in T2DM patients. In the group consuming high-dosage of probiotic, insulin sensitivity index (ISI) was high [97]. Table 4 provides an overview of important studies demonstrating the effect of probiotics on human subjects with type 2 diabetes mellitus.

ProbioticsModel typeMechanism of actionMajor findingsReferences
Bifidobacterium animalis A628 type II diabetic patientsSignificant decrease in fasting blood glucose, serum content of total cholesterol, the cardiovascular risk index (TC/HDL-C), the pro-inflammatory cytokines (IL-6, MCP-1) and adipokines (adiponectin, resistin, lipocalin-2, adipsin). Myokines (irisin, osteocrin) increased significantly, indicating possible improvement in skeletal muscle functionProbiotic camel milk powder twice a day for a consecutive four weeks can significantly decrease fasting blood glucose of type 2 diabetic patients[98]
probiotic supplements including Bifidobacterium bifidum 2 × 109, Lactobacillus casei 2 × 109, Lactobacillus acidophilus 2 × 109 CFU/day (n = 30)60 diabetic patients with CHD, aged 40–85 yearsDecreasing inflammatory cytokines and suppressing the nuclear factor-κB pathway, their impact on gene expression and the activation of gut microbiota short-chain fatty acids (SCFA)-hormone axisProbiotic supplementation for 12 weeks had beneficial effects on glycemic control, HDL-cholesterol, total-/HDL-cholesterol ratio, biomarkers of inflammation and oxidative stress in diabetic patients with CHD[99]
Lactobacillus reuteri68 T2DM patientsL. reuteri may influence changes in intestinal flora, which may lead to different outcomes after probiotic intake.Significant reductions in HbA1c and serum cholesterol
Bifidobacterium spp. were significantly increased
[96]
Symbiter53 patientsSignificant reduction of HOMA-IR from 6.85 ± 0.76 to 5.13 ± 0.49 (p = 0.047), but remained static in the placebo group. With respect to our secondary outcomes, HbA1cModestly improved insulin resistance[100]
Ecologic®Barrierpatients with type 2 diabetes mellitussignificant decrease in circulating levels of endotoxin by almost 70% over six months, as well as glucose (38%), insulin (38%), HOMA-IR (64%), triglycerides (48%), total cholesterol (19%), total/HDL-cholesterol ratio (19%), TNF-α (67%), IL-6 (77%), CRP (53%), resistin (53%), and a significant increase in adiponectin (72%) as compared with baselinemultistrain probiotics is a promising adjuvant antidiabetes therapy[93]

Table 4.

Overview of important studies demonstrating the effect of probiotics on human subjects with type 2 diabetes mellitus.

A 12-week probiotic therapy that comprised a multistrain probiotic was administered to 101 adults with T2DM. This intervention revealed that the probiotic intake lowers insulin resistance, fasting blood glucose, and HbA1C levels [101]. In an randomized controlled trial (RCT) by Palacios et al., patients with prediabetes and T2DM were enrolled to examine the outcomes of a probiotic multistrain. The only thing that separated the intervention and placebo groups was an increase in butyrate levels. It is noteworthy that those taking both metformin and a probiotic had decreased levels of insulin resistance, FBG, and HbA1c [102]. In a trial utilizing a single-strain probiotic (108 CFU/day of Lactobacillus casei for eight weeks), Khalili et al. discovered a decrease in FBG, insulin concentration, and insulin resistance [103].

Advertisement

4. Probiotic interventions to ameliorate gestational diabetes

Most of RCTs investigating the therapeutic benefits of probiotic supplementation in female GDM patients have been carried out in Iran; each study used a unique combination of microorganisms and examined a variety of outcomes in addition to glycemia such as gestational weight change [104], lipid profile [105], and inflammation [106]. Fasting blood sugar levels and insulin resistance dramatically decreased in the probiotic group in all these studies. Probiotics also decreased gestational weight gain, serum very-low-density lipoprotein (VLDL) cholesterol, and triglyceride levels [104, 105, 106]. Figure 4 shows an overview of potential mechanisms of probiotics against type 1 diabetes.

Figure 4.

Potential mechanisms of probiotics against gestational diabetes.

A recent study that randomly assigned GDM patients to receive probiotics (109 colony-forming units (CFU) per day of Bifidobacterium bifidum and Lactobacillus acidophilus or a placebo for four weeks also found significant improvement in glucose metabolism in the probiotic group, including fasting glucose, insulin, and HOMA-IR. An RCT conducted in Ireland randomized 149 women (GDM sufferers) to receive either a probiotic (Lactobacillus salivarius, 109 CFU per day) or a placebo, and the results showed no change between the two groups except for total cholesterol [107]. There was a considerable decrease in insulin resistance, which seemed to be primarily related to the species Bifidobacterium [108]. According to the findings, bigger, longer-term studies comparing various probiotic strains were required.

A modest number of RCTs have looked at probiotic supplementation’s potential to stop GDM. In the Finnish “Probiotics and Pregnancy Outcome Study, “ pregnant women were randomly assigned to receive dietary advice with probiotic supplementation (1010 CFU per day of Lactobacillus rhamnosus and Bifidobacterium lactis), dietary advice alone, or a placebo. In the probiotic group, the rate of GDM was much lower as compared to the other groups. There were no abnormalities in fetal development [109]. Table 5 provides an overview of important studies demonstrating the effect of probiotics on human subjects with gestational diabetes. Recently, a probiotic intervention study for women at risk of GDM in New Zealand has been carried out. At 14–16 weeks of gestation, the scientists randomly assigned women to take either a probiotic (Lactobacillus rhamnosus, 6 × 109 CFU per day) or a placebo. They also noticed that the probiotic intervention dramatically reduced the occurrence of GDM [110]. Even though the results are encouraging, further research is required to decide if probiotic supplements should be widely utilized in early pregnancy to prevent GDM.

ProbioticsModel typeMajor findingsReferences
Lactobacillus rhamnosus GG and Bifidobacterium lactis Bb12Pregnant women; no chronic diseases apart from allergic diseases; less than 17 gwReducing prevalence of GDM[109]
Lactobacillus salivarius UCC118Women with GDMNo significant effect on the incidence of GDM[107]
L. rhamnosus HN001Pregnant women with a personal or partner history of atopic diseaseReducing the prevalence of GDM[110]
myo-inositol 2 g + B. lactis and L. rhamnosusMexican women with three or more risk factors for developing GDMReducing the prevalence of GDM[111]

Table 5.

Overview of important studies demonstrating the effect of probiotics on human subjects with gestational diabetes.

Advertisement

5. Conclusion

Our hypothesis is that the manipulation of the intestinal flora by probiotics may be useful for the prevention and treatment of diabetes. Experimental and clinical trials have shown the significant potential of probiotic strains in the management of diabetes. Probiotics may increase insulin signaling molecules and insulin sensitivity, reduce inflammation and inflammatory cytokines, suppress the nuclear factor-κB pathway, activate gut microbiota short-chain fatty acids (SCFAs)-hormone axis, and enhance the barrier property of the gastrointestinal mucosa by altering the intestinal flora. The studies discussed in this chapter provide insights into the impact of probiotics on diabetes, although further investigation is required to clarify the molecular processes involved in the regulation of intestinal flora by probiotic administration and their effects on the onset of diabetes.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Blair M. Diabetes mellitus review. Urologic Nursing. 2016;36:27-36
  2. 2. Papatheodorou K, Banach M, Bekiari E, Rizzo M, Edmonds M. Complications of diabetes 2017. Journal Diabetes Research. 2018;2018:4. Article ID 3086167. DOI: 10.1155/2018/3086167
  3. 3. Delahanty LM, Halford BN. The role of diet behaviors in achieving improved glycemic control in intensively treated patients in the diabetes control and complications trial. Diabetes Care. 1993;16:1453-1458. DOI: 10.2337/DIACARE.16.11.1453
  4. 4. Papatheodorou K, Papanas N, Banach M, Papazoglou D, Edmonds M. Complications of diabetes 2016. Journal Diabetes Research. 2016;2016:4. Article ID 3086167. DOI: 10.1155/2016/6989453
  5. 5. World Health Organization. World Malaria Report 2015. Geneva, Switzerland: World Health Organization; 2016
  6. 6. Seuring T, Archangelidi O, Suhrcke M. The economic costs of type 2 diabetes: A global systematic review. PharmacoEconomics. 2015;33:811-831. DOI: 10.1007/S40273-015-0268-9
  7. 7. Ramachandran A, Snehalatha C. Current scenario of diabetes in India. Journal of Diabetes. 2009;1:18-28. DOI: 10.1111/J.1753-0407.2008.00004.X
  8. 8. Ramachandran A, Snehalatha C, Mary S, Mukesh B, Bhaskar AD, Vijay V. The Indian Diabetes Prevention Programme shows that lifestyle modification and metformin prevent type 2 diabetes in Asian Indian subjects with impaired glucose tolerance (IDPP-1). Diabetologia. 2006;49:289-297. DOI: 10.1007/S00125-005-0097-Z
  9. 9. Association AD. Classification and diagnosis of diabetes. Diabetes Care. 2017;40:S11-S24. DOI: 10.2337/DC17-S005
  10. 10. Association AD. Classification and diagnosis of diabetes. Diabetes Care. 2015;38:S8-S16. DOI: 10.2337/DC15-S005
  11. 11. Association AD. Diagnosis and classification of diabetes mellitus. Diabetes Care. 2009;32:S62-S67. DOI: 10.2337/DC09-S062
  12. 12. Bhat IA, Kabeer SW, Reza MI, Mir RH, Dar MO. AdipoRon: A novel insulin sensitizer in various complications and the underlying mechanisms: A review. Current Molecular Pharmacology. 2020;13:94-107. DOI: 10.2174/1874467212666191022102800
  13. 13. Lamichhane S, Ahonen L, Dyrlund TS, Siljander H, Hyöty H, Ilonen J, et al. A longitudinal plasma lipidomics dataset from children who developed islet autoimmunity and type 1 diabetes. Scientific Data. 2018;5:1-9. DOI: 10.1038/sdata.2018.250
  14. 14. Rewers M, Hyöty H, Lernmark Å, Hagopian W, She JX, Schatz D, et al. The environmental determinants of diabetes in the young (TEDDY) study: 2018 update. Current Diabetes Reports. 2018;2018:18. DOI: 10.1007/S11892-018-1113-2
  15. 15. Battaglia M, Atkinson MA. The streetlight effect in type 1 diabetes. Diabetes. 2015;64:1081-1090. DOI: 10.2337/DB14-1208
  16. 16. Cho NH, Shaw JE, Karuranga S, Huang Y, da Rocha Fernandes JD, Ohlrogge AW, et al. IDF Diabetes Atlas: Global estimates of diabetes prevalence for 2017 and projections for 2045. Diabetes Research and Clinical Practice. 2018;138:271-281. DOI: 10.1016/J.DIABRES.2018.02.023
  17. 17. Lyssenko V, Jonsson A, Almgren P, Pulizzi N, Isomaa B, Tuomi T, et al. Clinical risk factors, DNA variants, and the development of type 2 diabetes. New England Journal of Medicine. 2008;359:2220-2232. DOI: 10.1056/nejmoa0801869
  18. 18. Bellamy L, Casas JP, Hingorani AD, Williams D. Type 2 diabetes mellitus after gestational diabetes: A systematic review and meta-analysis. Lancet. 2009;373:1773-1779. DOI: 10.1016/S0140-6736(09)60731-5
  19. 19. Kubo A, Ferrara A, Windham GC, Greenspan LC, Deardorff J, Hiatt RA, et al. Maternal hyperglycemia during pregnancy predicts adiposity of the offspring. Diabetes Care. 2014;37:2996-3002. DOI: 10.2337/DC14-1438
  20. 20. Asemi Z, Tabassi Z, Samimi M, Fahiminejad T, Esmaillzadeh A. Favourable effects of the dietary approaches to stop hypertension diet on glucose tolerance and lipid profiles in gestational diabetes: A randomised clinical trial. The British Journal of Nutrition. 2013;109:2024-2030. DOI: 10.1017/S0007114512004242
  21. 21. Valdes AM, Walter J, Segal E, Spector TD. Role of the gut microbiota in nutrition and health. BMJ. 2018;361:36-44. DOI: 10.1136/BMJ.K2179
  22. 22. Li W-Z, Stirling K, Yang J-J, Zhang L. Gut microbiota and diabetes: From correlation to causality and mechanism. 2020;11:293-308. DOI: 10.4239/WJD.V11.I7.293
  23. 23. Larsen N, Vogensen FK, van den Berg FWJ, Nielsen DS, Andreasen AS, Pedersen BK, et al. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS One. 2010;2010:5. DOI: 10.1371/JOURNAL.PONE.0009085
  24. 24. Lê KA, Li Y, Xu X, Yang W, Liu T, Zhao X, et al. Alterations in fecal Lactobacillus and Bifidobacterium species in type 2 diabetic patients in southern China population. Frontiers in Physiology. 2013;2013:3. DOI: 10.3389/FPHYS.2012.00496
  25. 25. Derrien M, Vaughan EE, Plugge CM, de Vos WM. Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. International Journal of Systematic and Evolutionary Microbiology. 2004;54:1469-1476. DOI: 10.1099/IJS.0.02873-0
  26. 26. Brown CT, Davis-Richardson AG, Giongo A, Gano KA, Crabb DB, Mukherjee N, et al. Gut microbiome metagenomics analysis suggests a functional model for the development of autoimmunity for type 1 diabetes. PLoS One. 2011;2011:6. DOI: 10.1371/JOURNAL.PONE.0025792
  27. 27. Karlsson CLJ, Önnerfält J, Xu J, Molin G, Ahrné S, Thorngren-Jerneck K. The microbiota of the gut in preschool children with normal and excessive body weight. Obesity (Silver Spring). 2012;20:2257-2261. DOI: 10.1038/OBY.2012.110
  28. 28. Wen L, Ley RE, Volchkov PY, Stranges PB, Avanesyan L, Stonebraker AC, et al. Innate immunity and intestinal microbiota in the development of type 1 diabetes. Nature. 2008;455:1109-1113. DOI: 10.1038/NATURE07336
  29. 29. Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes. 2007;56:1761-1772. DOI: 10.2337/DB06-1491
  30. 30. Padwal R, Majumdar SR, Johnson JA, Varney J, McAlister FA. A systematic review of drug therapy to delay or prevent type 2 diabetes. Diabetes Care. 2005;28:736-744. DOI: 10.2337/DIACARE.28.3.736
  31. 31. Kumar A, Prajapati B, Pramanik J. Prebiotics as adjunctive therapy in diabetes: A review prebiotics in diabetes. Current Nutraceuticals. 2022;2022:03. DOI: 10.2174/2665978603666220818085437
  32. 32. Walker KZ, O’Dea K, Gomez M, Girgis S, Colagiuri R. Diet and exercise in the prevention of diabetes. Journal of Human Nutrition and Dietetics. 2010;23:344-352. DOI: 10.1111/J.1365-277X.2010.01061.X
  33. 33. Norris SL, Zhang X, Avenell A, Gregg E, Bowman B, Schmid CH, et al. Long-term effectiveness of weight-loss interventions in adults with pre-diabetes: A review. American Journal of Preventive Medicine. 2005;28:126-139. DOI: 10.1016/J.AMEPRE.2004.08.006
  34. 34. Norris SL, Zhang X, Avenell A, Gregg E, Brown T, Schmid CH, et al. Long-term non-pharmacological weight loss interventions for adults with type 2 diabetes mellitus. Cochrane Database of Systematic Reviews. 2005;2010(2):CD004095. DOI: 10.1002/14651858.CD004095.PUB2/INFORMATION/EN
  35. 35. Diabetes Management. Community Health Workers | the Community Guide. n.d. https://www.thecommunityguide.org/findings/diabetes-management-interventions-engaging-community-health-workers.html [Accessed December 9, 2022]
  36. 36. Gismondo MR, Drago L, Lombardi A. Review of probiotics available to modify gastrointestinal flora. International Journal of Antimicrobial Agents. 1999;12:287-292. DOI: 10.1016/S0924-8579(99)00050-3
  37. 37. Lilly DM, Stillwell RH. Probiotics: Growth-promoting factors produced by microorganisms. Science. 1979;1965(147):747-748. DOI: 10.1126/SCIENCE.147.3659.747
  38. 38. Parker RB. Probiotics, the other half of the antibiotic story. Animal Nutrition and Health. 1974;29:4-8
  39. 39. Reid G, Sanders ME, Gaskins HR, Gibson GR, Mercenier A, Rastall R, et al. New scientific paradigms for probiotics and prebiotics. Journal of Clinical Gastroenterology. 2003;37:105-118. DOI: 10.1097/00004836-200308000-00004
  40. 40. Hegar B, Waspada MI, Gunardi H, Vandenplas Y. A double blind randomized trial showing probiotics to be ineffective in acute Diarrhea in Indonesian children. Indian Journal of Pediatrics. 2015;82:410-414. DOI: 10.1007/S12098-014-1408-5
  41. 41. Cammarota M, de Rosa M, Stellavato A, Lamberti M, Marzaioli I, Giuliano M. In vitro evaluation of Lactobacillus plantarum DSMZ 12028 as a probiotic: Emphasis on innate immunity. International Journal of Food Microbiology. 2009;135:90-98. DOI: 10.1016/J.IJFOODMICRO.2009.08.022
  42. 42. Amara AA, Shibl A. Role of probiotics in health improvement, infection control and disease treatment and management. Saudi Pharmaceutical Journal: SPJ. 2015;23:107. DOI: 10.1016/J.JSPS.2013.07.001
  43. 43. Bourdichon F, Casaregola S, Farrokh C, Frisvad JC, Gerds ML, Hammes WP, et al. Food fermentations: Microorganisms with technological beneficial use. International Journal of Food Microbiology. 2012;154:87-97. DOI: 10.1016/J.IJFOODMICRO.2011.12.030
  44. 44. Metchnikoff I. The Prolongation of Life: Optimistic Studies. United States: Springer Publishing Company; 2004
  45. 45. Sanders ME, Merenstein D, Merrifield CA, Hutkins R. Probiotics for human use. Nutrition Bulletin. 2018;43:212-225. DOI: 10.1111/NBU.12334
  46. 46. Pawar R, Zambare V, Nabar B. Comparative assessment of antibiotic resistance in lactic acid Bacteria isolated from healthy human adult and infant Feces. Nepal Journal of Biotechnology. 2020;8:69-75. DOI: 10.3126/NJB.V8I2.31893
  47. 47. Choudhari G, Zambare V, Pawar R. Screening, isolation and characterization of probiotically safe lactic acid bacteria from human faeces for biofilm formation. International Journal of Research in BioSciences. 2018;7:10-18
  48. 48. Topping DL, Clifton PM. Short-chain fatty acids and human colonic function: Roles of resistant starch and nonstarch polysaccharides. Physiological Reviews. 2001;81:1031-1064. DOI: 10.1152/PHYSREV.2001.81.3.1031
  49. 49. Versalovic J. The human microbiome and probiotics: Implications for pediatrics. Annals of Nutrition & Metabolism. 2013;63(Suppl. 2):42-52. DOI: 10.1159/000354899
  50. 50. Pawar R, Dhawal P, Nabar B, Barve S, Zambare V. Mechanisms and applications of probiotics in healthcare industry. Biotechnology in Healthcare. 2022;2022:225-257. DOI: 10.1016/B978-0-323-90042-3.00002-5
  51. 51. Kristensen NB, Bryrup T, Allin KH, Nielsen T, Hansen TH, Pedersen O. Alterations in fecal microbiota composition by probiotic supplementation in healthy adults: A systematic review of randomized controlled trials. Genome Medicine. 2016;2016:8. DOI: 10.1186/S13073-016-0300-5
  52. 52. Sanders ME, Heimbach JT, Pot B, Tancredi D, Lenoir-Wijnkoop I, Lähteenmäki-Uutela A, et al. Health claims substantiation for probiotic and prebiotic products. Gut Microbes. 2011;2:127-133. DOI: 10.4161/GMIC.2.3.16174
  53. 53. Nagpal R, Wang S, Ahmadi S, Hayes J, Gagliano J, Subashchandrabose S, et al. Human-origin probiotic cocktail increases short-chain fatty acid production via modulation of mice and human gut microbiome. Scientific Reports. 2018;2018:8. DOI: 10.1038/S41598-018-30114-4
  54. 54. Ang Z, Ding JL. GPR41 and GPR43 in obesity and inflammation - protective or causative? Frontiers in Immunology. 2016;2016:7. DOI: 10.3389/FIMMU.2016.00028
  55. 55. Psichas A, Sleeth ML, Murphy KG, Brooks L, Bewick GA, Hanyaloglu AC, et al. The short chain fatty acid propionate stimulates GLP-1 and PYY secretion via free fatty acid receptor 2 in rodents. International Journal of Obesity. 2015;39:424-429. DOI: 10.1038/IJO.2014.153
  56. 56. Christiansen CB, Gabe MBN, Svendsen B, Dragsted LO, Rosenkilde MM, Holst JJ. The impact of short-chain fatty acids on GLP-1 and PYY secretion from the isolated perfused rat colon. American Journal of Physiology. Gastrointestinal and Liver Physiology. 2018;315:G53-G65. DOI: 10.1152/AJPGI.00346.2017
  57. 57. Abdelazez A, Abdelmotaal H, Evivie SE, Melak S, Jia FF, Khoso MH, et al. Screening potential probiotic characteristics of lactobacillus brevis strains in vitro and intervention effect on type I diabetes in vivo. Biomedical Research International. 2018;2018:20. Article ID 7356173. DOI: 10.1155/2018/7356173
  58. 58. Dolpady J, Sorini C, di Pietro C, Cosorich I, Ferrarese R, Saita D, et al. Oral probiotic VSL#3 prevents autoimmune diabetes by modulating microbiota and promoting indoleamine 2,3-dioxygenase-enriched tolerogenic intestinal environment. Journal Diabetes Research. 2016;2016:12. Article ID 7569431. DOI: 10.1155/2016/7569431
  59. 59. van Endert P, Mauvais FX, Diana J. Beta cell antigens in type 1 diabetes: Triggers in pathogenesis and therapeutic targets. F1000Res. 2016;2016:5. DOI: 10.12688/F1000RESEARCH.7411.1/DOI
  60. 60. Takiishi T, Korf H, van Belle TL, Robert S, Grieco FA, Caluwaerts S, et al. Reversal of autoimmune diabetes by restoration of antigen-specific tolerance using genetically modified Lactococcus lactis in mice. The Journal of Clinical Investigation. 2012;122:1717-1725. DOI: 10.1172/JCI60530
  61. 61. Calcinaro F, Dionisi S, Marinaro M, Candeloro P, Bonato V, Marzotti S, 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. DOI: 10.1007/S00125-005-1831-2
  62. 62. Yadav R, Dey DK, Vij R, Meena S, Kapila R, Kapila S. Evaluation of anti-diabetic attributes of Lactobacillus rhamnosus MTCC: 5957, Lactobacillus rhamnosus MTCC: 5897 and Lactobacillus fermentum MTCC: 5898 in streptozotocin induced diabetic rats. Microbial Pathogenesis. 2018;125:454-462. DOI: 10.1016/J.MICPATH.2018.10.015
  63. 63. Le TKC, Hosaka T, Nguyen TT, Kassu A, Dang TO, Tran HB, et al. Bifidobacterium species lower serum glucose, increase expressions of insulin signaling proteins, and improve adipokine profile in diabetic mice. Biomedical Research. 2015;36:63-70. DOI: 10.2220/BIOMEDRES.36.63
  64. 64. Zhang J, Motyl KJ, Irwin R, MacDougald OA, Britton RA, McCabe LR. Loss of bone and Wnt10b expression in male type 1 diabetic mice is blocked by the probiotic Lactobacillus reuteri. Endocrinology. 2015;156:3169-3182. DOI: 10.1210/EN.2015-1308
  65. 65. Wei SH, Chen YP, Chen MJ. Selecting probiotics with the abilities of enhancing GLP-1 to mitigate the progression of type 1 diabetes in vitro and in vivo. Journal of Functional Foods. 2015;18:473-486. DOI: 10.1016/J.JFF.2015.08.016
  66. 66. Valladares R, Sankar D, Li N, Williams E, Lai KK, Abdelgeliel AS, et al. Lactobacillus johnsonii N6.2 mitigates the development of type 1 diabetes in BB-DP rats. PLoS One. 2010;2010:5. DOI: 10.1371/JOURNAL.PONE.0010507
  67. 67. Lau K, Benitez P, Ardissone A, Wilson TD, Collins EL, Lorca G, et al. Inhibition of type 1 diabetes correlated to a Lactobacillus johnsonii N6.2-mediated Th17 bias. Journal of Immunology. 2011;186:3538-3546. DOI: 10.4049/JIMMUNOL.1001864
  68. 68. Bejar W, Hamden K, Salah R, Chouayekh H. Lactobacillus plantarum TN627 significantly reduces complications of alloxan-induced diabetes in rats. Anaerobe. 2013;24:4-11. DOI: 10.1016/J.ANAEROBE.2013.08.006
  69. 69. Vatanen T, Franzosa EA, Schwager R, Tripathi S, Arthur TD, Vehik K, et al. The human gut microbiome in early-onset type 1 diabetes from the TEDDY study. Nature. 2018;562:589-594. DOI: 10.1038/S41586-018-0620-2
  70. 70. Uusitalo U, Liu X, Yang J, Aronsson CA, Hummel S, Butterworth M, et al. Association of Early Exposure of probiotics and islet autoimmunity in the TEDDY study. JAMA Pediatrics. 2016;170:20-28. DOI: 10.1001/JAMAPEDIATRICS.2015.2757
  71. 71. Marcial GE, Ford AL, Haller MJ, Gezan SA, Harrison NA, Cai D, et al. Lactobacillus johnsonii N6.2 modulates the host immune responses: A double-blind, randomized trial in healthy adults. Frontiers in Immunology. 2017;2017:8. DOI: 10.3389/FIMMU.2017.00655
  72. 72. Weir GC, Bonner-Weir S. Dreams for type 1 diabetes: Shutting off autoimmunity and stimulating beta-cell regeneration. Endocrinology. 2010;151:2971-2973. DOI: 10.1210/EN.2010-0538
  73. 73. Yadav H, Lee JH, Lloyd J, Walter P, Rane SG. Beneficial metabolic effects of a probiotic via butyrate-induced GLP-1 hormone secretion. The Journal of Biological Chemistry. 2013;288:25088-25097. DOI: 10.1074/JBC.M113.452516
  74. 74. Burrows MP, Volchkov P, Kobayashi KS, Chervonsky A, v., Medzhitov R. Microbiota regulates type 1 diabetes through toll-like receptors. Proceedings of the National Academy of Sciences of the United States of America. 2015;112:9973-9977. DOI: 10.1073/PNAS.1508740112
  75. 75. Groele L, Szajewska H, Szypowska A. Effects of Lactobacillus rhamnosus GG and Bifidobacterium lactis Bb12 on beta-cell function in children with newly diagnosed type 1 diabetes: Protocol of a randomised controlled trial. BMJ Open. 2017;2017:7. DOI: 10.1136/BMJOPEN-2017-017178
  76. 76. Ahola AJ, Harjutsalo V, Forsblom C, Freese R, Makimattila S, Groop P. The self-reported use of probiotics is associated with better glycaemic control and lower odds of metabolic syndrome and its components in type 1 diabetes. Journal of Probiotics Health. 2017;2017:05. DOI: 10.4172/2329-8901.1000188
  77. 77. Yang J, Tamura RN, Uusitalo UM, Aronsson CA, Silvis K, Riikonen A, et al. Vitamin D and probiotics supplement use in young children with genetic risk for type 1 diabetes. European Journal of Clinical Nutrition. 2017;71:1449-1454. DOI: 10.1038/EJCN.2017.140
  78. 78. Lee YK, Margolles A, Mayo B, Ruas-Madiedo P, Gueimonde M, de los Reyes-Gavilán C, et al. Probiotic Microorganisms. Handbook of Probiotics and Prebiotics. Second ed. New York, United States. 2008. pp. 1-176. DOI: 10.1002/9780470432624.CH1
  79. 79. Li X, Wang N, Yin B, Fang D, Jiang T, Fang S, et al. Effects of Lactobacillus plantarum CCFM0236 on hyperglycaemia and insulin resistance in high-fat and streptozotocin-induced type 2 diabetic mice. Journal of Applied Microbiology. 2016;121:1727-1736. DOI: 10.1111/JAM.13276
  80. 80. Lee E, Jung SR, Lee SY, Lee NK, Paik HD, Lim S. Lactobacillus plantarum strain Ln4 attenuates diet-induced obesity, insulin resistance, and changes in hepatic mRNA levels associated with glucose and lipid metabolism. Nutrients. 2018;2018:10. DOI: 10.3390/NU10050643
  81. 81. Balakumar M, Prabhu D, Sathishkumar C, Prabu P, Rokana N, Kumar R, et al. Improvement in glucose tolerance and insulin sensitivity by probiotic strains of Indian gut origin in high-fat diet-fed C57BL/6J mice. European Journal of Nutrition. 2018;57:279-295. DOI: 10.1007/S00394-016-1317-7
  82. 82. Dang F, Jiang Y, Pan R, Zhou Y, Wu S, Wang R, et al. Administration of Lactobacillus paracasei ameliorates type 2 diabetes in mice. Food & Function. 2018;9:3630-3639. DOI: 10.1039/C8FO00081F
  83. 83. Memarrast F, Ghafouri-Fard S, Kolivand S, Jafary-Nodooshan S, Neyazi N, Sadroddiny E, et al. Comparative evaluation of probiotics effects on plasma glucose, lipid, and insulin levels in streptozotocin-induced diabetic rats. Diabetes/Metabolism Research and Reviews. 2017;2017:33. DOI: 10.1002/DMRR.2912
  84. 84. Lin Y, Ren Y, Zhang Y, Zhou J, Zhou F, Zhao Q, et al. Protective role of nano-selenium-enriched Bifidobacterium longum in delaying the onset of streptozotocin-induced diabetes. Royal Society Open Science. 2018;2018:5. DOI: 10.1098/RSOS.181156
  85. 85. Sakai T, Taki T, Nakamoto A, Shuto E, Tsutsumi R, Toshimitsu T, et al. Lactobacillus plantarum OLL2712 regulates glucose metabolism in C57BL/6 mice fed a high-fat diet. Journal of Nutritional Science and Vitaminology (Tokyo). 2013;59:144-147. DOI: 10.3177/JNSV.59.144
  86. 86. Wang G, Li X, Zhao J, Zhang H, Chen W. Lactobacillus casei CCFM419 attenuates type 2 diabetes via a gut microbiota dependent mechanism. Food & Function. 2017;8:3155-3164. DOI: 10.1039/C7FO00593H
  87. 87. 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. Molecular Nutrition & Food Research. 2019;2019:63. DOI: 10.1002/MNFR.201900457
  88. 88. Jia L, Li D, Feng N, Shamoon M, Sun Z, Ding L, et al. Anti-diabetic effects of Clostridium butyricum CGMCC0313.1 through promoting the growth of gut butyrate-producing Bacteria in type 2 diabetic mice. Scientific Reports. 2017:7. DOI: 10.1038/S41598-017-07335-0
  89. 89. Manaer T, Yu L, Nabi XH, Dilidaxi D, Liu L, Sailike J. The beneficial effects of the composite probiotics from camel milk on glucose and lipid metabolism, liver and renal function and gut microbiota in db/db mice. BMC Complementary Medicine Therapy. 2021;21:1-13. DOI: 10.1186/S12906-021-03303-4/FIGURES/6
  90. 90. Zhao D, Zhu H, Gao F, Qian Z, Mao W, Yin Y, et al. Antidiabetic effects of selenium-enriched Bifidobacterium longum DD98 in type 2 diabetes model of mice. Food & Function. 2020;11:6528-6541. DOI: 10.1039/D0FO00180E
  91. 91. 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. Journal of Functional Foods. 2020;67:103843. DOI: 10.1016/J.JFF.2020.103843
  92. 92. Sabico S, Al-Mashharawi A, Al-Daghri NM, Yakout S, Alnaami AM, Alokail MS, et al. Effects of a multi-strain probiotic supplement for 12 weeks in circulating endotoxin levels and cardiometabolic profiles of medication naïve T2DM patients: A randomized clinical trial. Journal of Translational Medicine. 2017;2017:15. DOI: 10.1186/S12967-017-1354-X
  93. 93. Sabico S, Al-Mashharawi A, Al-Daghri NM, Wani K, Amer OE, Hussain DS, et al. 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. Clinical Nutrition. 2019;38:1561-1569. DOI: 10.1016/J.CLNU.2018.08.009
  94. 94. Karczewski J, Troost FJ, Konings I, Dekker J, Kleerebezem M, Brummer RJM, et al. Regulation of human epithelial tight junction proteins by Lactobacillus plantarum in vivo and protective effects on the epithelial barrier. American Journal of Physiology. Gastrointestinal and Liver Physiology. 2010;298:851-859. DOI: 10.1152/AJPGI.00327.2009/ASSET/IMAGES/LARGE/ZH30051056450004.JPEG
  95. 95. Hummel S, Veltman K, Cichon C, Sonnenborn U, Schmidt MA. Differential targeting of the E-cadherin/β-catenin complex by gram-positive probiotic lactobacilli improves epithelial barrier function. Applied and Environmental Microbiology. 2012;78:1140-1147. DOI: 10.1128/AEM.06983-11/ASSET/F277D377-DD28-48F7-8448-623CFA15F5AD/ASSETS/GRAPHIC/ZAM9991029470006.JPEG
  96. 96. Hsieh MC, Tsai WH, Jheng YP, Su SL, Wang SY, Lin CC, et al. The beneficial effects of Lactobacillus reuteri ADR-1 or ADR-3 consumption on type 2 diabetes mellitus: A randomized, double-blinded, placebo-controlled trial. Scientific Reports. 2018;8:1-11. DOI: 10.1038/s41598-018-35014-1
  97. 97. Mobini R, Tremaroli V, Ståhlman M, Karlsson F, Levin M, Ljungberg M, et al. Metabolic effects of Lactobacillus reuteri DSM 17938 in people with type 2 diabetes: A randomized controlled trial. Diabetes, Obesity & Metabolism. 2017;19:579-589. DOI: 10.1111/DOM.12861
  98. 98. B F, M Z, L D, X Z, F R, S G. Probiotic camel milk powder improves glycemic control, dyslipidemia, adipose tissue and skeletal muscle function in T2DM patients: A randomized trial. 2020. 10.21203/RS.3.RS-34115/V1
  99. 99. Raygan F, Rezavandi Z, Bahmani F, Ostadmohammadi V, Mansournia MA, Tajabadi-Ebrahimi M, et al. The effects of probiotic supplementation on metabolic status in type 2 diabetic patients with coronary heart disease IRCT2017082733941N5 IRCT. Diabetology and Metabolic Syndrome. 2018;10:1-7. DOI: 10.1186/S13098-018-0353-2/TABLES/3
  100. 100. 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 & Metabolic Syndrome: Clinical Research & Reviews. 2018;12:617-624. DOI: 10.1016/J.DSX.2018.04.015
  101. 101. Firouzi S, Majid HA, Ismail A, Kamaruddin NA, Barakatun-Nisak MY. Effect of multi-strain probiotics (multi-strain microbial cell preparation) on glycemic control and other diabetes-related outcomes in people with type 2 diabetes: A randomized controlled trial. European Journal of Nutrition. 2016;56:1535-1550. DOI: 10.1007/S00394-016-1199-8
  102. 102. Palacios T, Vitetta L, Coulson S, Madigan CD, Lam YY, Manuel R, et al. Targeting the intestinal microbiota to prevent Type 2 diabetes and enhance the effect of metformin on glycaemia: A Randomised Controlled Pilot Study. Nutrients. 2020;12:2041. DOI: 10.3390/NU12072041
  103. 103. Khalili L, Alipour B, Jafar-Abadi MA, Faraji I, Hassanalilou T, Abbasi MM, et al. 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. Iranian Biomedical Journal. 2019;23:68-77. DOI: 10.29252/.23.1.68
  104. 104. Dolatkhah N, Hajifaraji M, Abbasalizadeh F, Aghamo-hammadzadeh N, Mehrabi Y, Abbasi MM. Is there a value for probiotic supplements in gestational diabetes mellitus? A randomized clinical trial. Journal of Health, Population, and Nutrition. 2015;33:1-8. DOI: 10.1186/S41043-015-0034-9/TABLES/3
  105. 105. Karamali M, Dadkhah F, Sadrkhanlou M, Jamilian M, Ahmadi S, Tajabadi-Ebrahimi M, et al. Effects of probiotic supplementation on glycaemic control and lipid profiles in gestational diabetes: A randomized, double-blind, placebo-controlled trial. Diabetes & Metabolism. 2016;42:234-241. DOI: 10.1016/J.DIABET.2016.04.009
  106. 106. Jafarnejad S, Saremi S, Jafarnejad F, Arab A. Effects of a multispecies probiotic mixture on Glycemic control and inflammatory status in women with gestational diabetes: A randomized controlled clinical trial. Journal of Nutrition Metabolism. 2016;2016. DOI: 10.1155/2016/5190846
  107. 107. Lindsay KL, Kennelly M, Culliton M, Smith T, Maguire OC, Shanahan F, et al. Probiotics in obese pregnancy do not reduce maternal fasting glucose: A double-blind, placebo-controlled, randomized trial (probiotics in pregnancy study). The American Journal of Clinical Nutrition. 2014;99:1432-1439. DOI: 10.3945/AJCN.113.079723
  108. 108. Taylor BL, Woodfall GE, Sheedy KE, O’Riley ML, Rainbow KA, Bramwell EL, et al. Effect of probiotics on metabolic outcomes in pregnant women with gestational diabetes: A systematic review and Meta-analysis of randomized controlled trials. Nutrients. 2017;2017:9. DOI: 10.3390/NU9050461
  109. 109. Luoto R, Laitinen K, Nermes M, Isolauri E. Impact of maternal probiotic-supplemented dietary counselling on pregnancy outcome and prenatal and postnatal growth: A double-blind, placebo-controlled study. The British Journal of Nutrition. 2010;103:1792-1799. DOI: 10.1017/S0007114509993898
  110. 110. Wickens KL, Barthow CA, Murphy R, Abels PR, Maude RM, Stone PR, et al. Early pregnancy probiotic supplementation with Lactobacillus rhamnosus HN001 may reduce the prevalence of gestational diabetes mellitus: A randomised controlled trial. British Journal of Nutrition. 2017;117:804-813. DOI: 10.1017/S0007114517000289
  111. 111. Reyes-Muñoz E, Espino Y, Sosa S, Flores-Robles CM, Arce-Sánchez L, Martínez-Cruz N, et al. Uso de mioinositol más Bifidobacterium lactis y Lactobacillus rhamnosus para la prevención de diabetes mellitus gestacional en mujeres mexicanas. Gaceta Médica de México. 2020;156:S51-S57. DOI: 10.24875/GMM.M20000438

Written By

Akash Kumar, Jhilam Pramanik, Nandani Goyal, Bhupendra G. Prajapati and Dimple Chauhan

Submitted: 07 January 2023 Reviewed: 02 February 2023 Published: 21 February 2024