Next Article in Journal
Evaluation of the Antifungal and Biochemical Activities of Fungicides and Biological Agents against Ginseng Sclerotinia Root Rot Caused by Sclerotinia nivalis
Next Article in Special Issue
Strain-Dependent Adhesion Variations of Shouchella clausii Isolated from Healthy Human Volunteers: A Study on Cell Surface Properties and Potential Probiotic Benefits
Previous Article in Journal
Impact of Graphene Layers on Genetic Expression and Regulation within Sulfate-Reducing Biofilms
Previous Article in Special Issue
Synbiotics as Treatment for Irritable Bowel Syndrome: A Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

New Insights into Chronic Pancreatitis: Potential Mechanisms Related to Probiotics

Department of Gastroenterology, State Key Laborotary of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Microorganisms 2024, 12(9), 1760; https://doi.org/10.3390/microorganisms12091760
Submission received: 8 August 2024 / Revised: 22 August 2024 / Accepted: 23 August 2024 / Published: 24 August 2024
(This article belongs to the Special Issue Probiotics: The Current State of Scientific Knowledge)

Abstract

:
Chronic pancreatitis is a progressive fibroinflammatory disorder with no currently satisfactory treatment. Emerging evidence suggests an association between gut microbial dysbiosis and chronic pancreatitis. Although direct causative evidence is lacking, it is hypothesized that the gut microbiota may play a pivotal role in modulating pancreatic function via the gut–pancreas axis. Thus, modulating the gut microbiota through the administration of probiotics or prebiotics may alleviate pancreatic disorders. In this review, we first propose the potential mechanisms by which specific probiotics or prebiotics may ameliorate chronic pancreatitis, including the alleviation of small intestinal bacterial overgrowth (SIBO), the facilitation of short-chain fatty acids’ (SCFAs) production, and the activation of glucagon-like peptide-1 receptors (GLP-1Rs) in the pancreas. Since there are currently no probiotics or prebiotics used for the treatment of chronic pancreatitis, we discuss research in other disease models that have used probiotics or prebiotics to modulate pancreatic endocrine and exocrine functions and prevent pancreatic fibrosis. This provides indirect evidence for their potential application in the treatment of chronic pancreatitis. We anticipate that this research will stimulate further investigation into the gut–pancreas axis and the potential therapeutic value of probiotics and prebiotics in chronic pancreatitis.

1. Introduction

Chronic pancreatitis (CP) is a progressive fibroinflammatory syndrome with an annual incidence from five to eight and a prevalence of 42–73 cases per 100,000 adults in the United States [1,2,3]. With repetitive episodes of inflammation, the pancreas is irreversibly replaced by fibrotic tissues, resulting in chronic abdominal pain, endocrine and exocrine insufficiency, a reduced quality of life, and a shorter life expectancy [4]. Current therapeutic approaches primarily focus on symptom alleviation and supportive care, rather than targeting the underlying pathophysiological mechanisms [5].
In recent years, accumulating evidence has highlighted the crucial role of the pancreas in regulating the gut microbiota and the reciprocal influence of the gut microbiota on pancreatic function, which indicates the presence of a bidirectional relationship referred to as the “gut–pancreas axis”. The gut microbiota play a pivotal role in this axis through their involvement in metabolism and nutrition, protection against pathogens, and immune system regulation [6]. Bidirectional alteration of the gut–pancreas axis has been observed in many pancreatic diseases, including CP (Figure 1) [7]. Regarding its role in the homeostasis of the gut–pancreas axis, microbiota-based treatments, such as probiotics and prebiotics, may offer effective therapeutic options for CP.
Probiotics, defined as live microorganisms that confer health benefits to the host, and prebiotics, non-digestible food components that selectively stimulate beneficial gut bacteria, have shown potential in managing various gastrointestinal and systemic disorders [8]. In the context of CP, although probiotics or prebiotics have been proposed as potential treatments [6,7], their efficacy has not yet been validated in animal models or clinical trials. Additionally, their possible mechanisms of action have not been thoroughly explored. This study aims to explore the potential of probiotics and prebiotics to enhance pancreatic function and treat CP by investigating their effects on the gut–pancreas axis. We will discuss the possible mechanisms that ameliorate CP, including endocrine and exocrine function improvement, inflammation reduction, and pancreatic fibrosis alleviation. The effects of probiotics and prebiotics on these targets and their feasibility as intervention methods are reviewed.
It is important to emphasize that, to date, there have been no animal experiments or clinical trials specifically investigating the use of probiotics or prebiotics for treating CP. The mechanisms we propose here are derived from the clinical manifestations of CP, such as endocrine and exocrine insufficiency and pancreatic inflammation. We base our hypothesis on evidence from other disease models, where probiotics and prebiotics have shown potential in improving pancreatic function and alleviating symptoms. However, whether these mechanisms are applicable to CP remains an open question that future animal and clinical studies need to address. It must be acknowledged that much work remains to be conducted before probiotics, including engineered strains, and prebiotics can be considered for the clinical treatment of CP. We hope to inspire new approaches and provide fresh insights into the potential treatment of CP.

2. Search Strategy

We conducted a comprehensive search in PubMed in July 2024. The search strategy involved the following main queries: (1) “(probiotics OR prebiotics OR synbiotics) AND (pancreatitis)”. Literature and reference screening were conducted to select potentially relevant articles. This approach provided a general overview of the current research landscape and the potential therapeutic mechanisms of probiotics and prebiotics in CP. After identifying these potential mechanisms of action, the following search queries were included: (2) (small intestine bacterial overgrowth) AND (chronic pancreatitis); (3) (small intestine bacterial overgrowth) AND (probiotics OR prebiotics OR synbiotics); (4) (short-chain fatty acid) AND (pancreas*); (5) (short-chain fatty acid) AND (probiotics OR prebiotics OR synbiotics); (6) (GLP-1) AND (pancreas*); and (7) (GLP-1) AND (probiotics OR prebiotics OR synbiotics). The literature search process of this review is shown in Figure 2.

3. Alleviation of Small Intestinal Bacterial Overgrowth

In a healthy small intestine, several defective mechanisms maintain a relatively sterile environment: gastric acid secretion, an intact ileocecal valve, intestinal motility, immunoglobulins in intestinal secretions, and the bacteriostatic properties of pancreatic and biliary secretions [9,10]. When these protective mechanisms are disrupted, small intestinal bacterial overgrowth syndrome (SIBO) can occur. SIBO is characterized by an excessive number of bacteria in the small bowel, leading to gastrointestinal symptoms such as bloating, abdominal distension, diarrhea, and nutrient deficiencies [11,12]. A systematic review found that SIBO is present in 38% of patients with CP [13]. Current evidence links SIBO in CP to diabetes mellitus, pancreatic exocrine insufficiency, and the severity of CP, with treatment often resulting in symptomatic improvement [13,14,15,16].
The standard treatment for SIBO involves antibiotics aimed at eradicating the bacteria in the small intestine [17]. However, with a combined normalization rate of 51% for antibiotics, about half of patients may remain symptomatic despite treatment [18]. This necessitates refined treatment strategies. Probiotics and prebiotics are believed to benefit SIBO by preventing the growth of pathogenic flora through direct competition and the production of bacteriocins [12]. Several randomized controlled trials have shown that adding probiotics to antibiotic therapy results in higher clinical remission rates [19,20,21]. In a randomized prospective pilot study of patients with SIBO and chronic abdominal distension, the group receiving a combination of probiotics (Lacticaseibacillus casei, Lactiplantibacillus plantarum, Streptococcus faecalis, and Bifidobacterium brevis) showed significantly better clinical improvements compared to the sole metronidazole group [22]. A systematic review concluded that, while probiotics are unable to prevent SIBO, they can effectively decontaminate SIBO and relieve abdominal pain [23]. Probiotics also aid in repairing and reconstructing the intestinal mucosa. In rats treated with probiotic formulations containing coconut oil and traces of peppermint–lemon–patchouli essential oil, researchers observed mitotic figures and the regression of the inflammatory response in the villus epithelium and crypts previously damaged by SIBO-induced gut dysbiosis [24].
Probiotic supplementation to reduce SIBO has been attempted in various diseases, including irritable bowel syndrome [25,26,27,28,29], hypothyroidism during pregnancy [30,31,32], systemic sclerosis [33], liver diseases [34,35,36], and gastric and colorectal cancer [37]. However, there is a lack of research evidence on the application of probiotics for SIBO in chronic pancreatitis. Further investigation is needed to explore the potential benefits of probiotics in alleviating SIBO in CP.

4. Facilitation of Short-Chain Fatty Acids’ Production

Short-chain fatty acids (SCFAs), primarily acetate, propionate, and butyrate, are produced via the fermentation of dietary fibers by the gut microbiota. They have significant effects on various tissues, including the pancreas. Sodium butyrate is capable of inhibiting histone deacetylases (HDACs), which are crucial in inflammation and fibrogenesis. Post-treatment with sodium butyrate significantly reduces the expression of α-smooth muscle actin, interleukin-1β, inducible nitric oxide synthase, and 3-nitrotyrosine, thereby alleviating L-arginine-induced pancreatic damage and fibrosis in rats [38]. SCFAs modulate pancreatic fibrosis by inhibiting macrophage infiltration and M2 phenotype switching [39]. SCFAs have also been confirmed to play an immunoregulatory and anti-inflammatory role. Cathelicidin-related antimicrobial peptide (CRAMP) is an immunoregulatory antimicrobial peptide that can be produced by acinar cells. It modulates the phenotypic switching of intrapancreatic macrophages and changes the production of transforming growth factor-β, thereby defending against inflammation. Research has revealed that the production of CRAMP is regulated by SCFAs produced by the gut microbiota [40]. Additionally, SCFAs, especially butyrate, exhibit anti-inflammatory effects by inhibiting the activation of NF-κB and HDACs [41,42,43,44]. SCFAs also act directly on acinar cells to stimulate secretion, similar to incretins, through increasing the cellular calcium concentration [45,46,47,48].
Extensive studies have investigated SCFAs’ effects on insulin secretion, acting as ligands to G-protein-coupled receptors (GPCRs), specifically, free fatty acid receptor-2 (FFA2, previously termed GPR43) and FFA3 (previously termed GPR41). These receptors are found in various human tissues, including gut enteroendocrine cells and pancreatic islets [49,50]. FFA2 and FFA3 receptors on enteroendocrine cells trigger GLP-1 secretion [49], which has multiple positive effects and will be discussed in the next part. The enhanced secretion of insulin after SCFA treatment has been reported in a number of studies and is thought to be associated with FFA2 and FFA3 receptors on β-cells, but contradicting evidence also exists in several studies [50,51]. Therefore, no clear consensus has been reached on the effect of SCFAs on the FFA2 and FFA3 receptors in pancreatic islets.
Patients with CP exhibit a reduced abundance of SCFA producers, such as Faecalibacterium and Fusicatenibacter [52]. There was a noticeable reduction in Faecalibacterium prausnitzii in healthy controls compared to CP non-diabetics and CP diabetics [53]. The depletion of SCFA-producing Gram-positive bacteria in CP is independent of toll-like receptor 4 (TLR4), but supplementing exogenous SCFAs ameliorates the condition [39]. These studies implicated the role of SCFAs in protecting the pancreatic function from damage caused by CP. Therefore, supplementing probiotics or prebiotics that contribute to SCFA production may offer a novel intervention for managing CP.
Both in vivo and in vitro studies confirm that probiotics can increase SCFA levels. The probiotics capable of producing SCFAs are summarized in Table 1. In an in vitro human gut model, an aqueous probiotic suspension containing L. plantarum, L. rhamnosus, L. acidophilus, and Enterococcus faecium exerted anti-inflammatory effects through increased SCFA production, especially butyrate [54].
Prebiotics also show potential as clinical targets by promoting the growth and activity of probiotics. Prebiotics, typically complex carbohydrates such as starch, pectin, xylan, and arabinogalactan, serve as substrates for bacterial fermentation, resulting in the production of SCFAs [59]. The metabolism of different polysaccharides is associated with the production of different SCFAs. For example, pectin metabolism leads to a proportional increase in acetate concentration, while starch fermentation significantly boosts butyrate production over other SCFAs [60,61]. Overall, the microbial hydrolysis of insoluble substrates can promote the biosynthesis of high concentrations of SCFAs, with about 60% presenting as acetate, while butyrate and propionate each account for approximately 20% of gastrointestinal SCFAs [62]. Colonic SCFAs increase in healthy humans after consuming inulin or arabinoxylan-oligosaccharides-enriched food [63,64]. Inulin supplementation elevates the abundance of butyrate-producing microbiota, including Bifidobacterium, Clostridium cluster IV, and Akkermansia muciniphila [65]. When supplemented with oligofructose or inulin as the sole energy source, cross-feeding interactions between bifidobacteria and butyrate-producing bacteria like Faecalibacterium prausnitzii are observed. These interactions may enhance the colon ecosystem and contribute to combined bifidogenic and butyrogenic effects [66,67].
In summary, the use of probiotics and prebiotics to produce SCFAs shows promise as a management technique for CP. This approach could help to modulate inflammation, fibrosis, and pancreatic function, offering a potential therapeutic avenue worth further exploration.

5. Activation of Glucagon-like Peptide 1 Receptors in the Pancreas

Glucagon-like peptide 1 (GLP-1) is released from gut enteroendocrine cells at low levels during fasting and increases significantly within minutes of food digestion. GLP-1 is a multifaceted hormone with broad pharmacological potential, including incretin-like activity, the stimulation of glucose-dependent insulin secretion, and the inhibition of glucagon secretion, food intake, and gastric emptying [68,69]. Although GLP-1 is currently only approved for the treatment of T2DM and obesity [70], its multiple effects on both the endocrine and exocrine pancreas suggest the potential for alleviating symptoms of CP and slowing disease progression.
The physiological importance of GLP-1R in β-cells has been well-established in animal studies. GLP-1 normalizes glucose tolerance and enhances glucose-dependent insulin secretion via GLP-1R in pancreatic β-cells [71]. In addition to directly inhibiting glucagon secretion by α-cells [72], GLP-1 stimulates somatostatin secretion by δ-cells, further reducing glucagon levels [73,74]. Beyond regulating the blood glucose through modulating the levels of insulin and glucagon, GLP-1 inhibits β-cell apoptosis, induces β-cell proliferation, and increases β-cell mass [75]. In diabetic mouse models, GLP-1R activation alleviates endoplasmic reticulum stress in β-cells via the cAMP-dependent enhancement of activating transcription factor 4 (ATF4) translation, promoting β-cell survival [76]. Although GLP-1R agonists can increase the β-cell mass in diabetic rodent models, this effect is modest and short-lived, with older rodents showing reduced responses [77,78,79]. Nevertheless, these drugs are believed to help prevent further losses of β-cell mass and function, especially if treatment begins early in disease progression. In baboons subjected to partial pancreatectomy and treated with the GLP-1R agonist exenatide, immunofluorescent staining revealed ductal cells co-expressing insulin, suggesting that exenatide might promote the differentiation of ductal cells into β-like cells [80].
While most GLP-1 research focuses on α- and β-cells in the endocrine pancreas, GLP-1 also affects the exocrine pancreas. GLP-1R is expressed in a significant proportion of pancreatic acinar cells, though at lower levels than in β cells [81,82,83]. In caerulein-induced experimental pancreatitis, GLP-1R agonists increased the pancreas weight and induced anti-inflammatory protein expression while reducing proinflammatory markers [84]. Preclinical studies show that GLP-1R activation increases the acinar cell mass and protein content via S6 phosphorylation, independent of DNA content or cell proliferation changes [85]. GLP-1 has been shown to induce amylase secretion in the pancreatic acini in a dose-independent manner, potentially alleviating the weakened exocrine digestive capacity in CP patients [81,86]. Nevertheless, further investigation is still needed to exclude the potential harm of GLP-1 overactivation of the exocrine pancreas. A T2DM population-based cohort study reported a 1.5-fold increased risk of any pancreatitis and a 2.0-fold increased risk of AP among incretin users, with no increased risk for CP [87]. Chronic GLP-1R agonist treatment has been associated with the activation of pancreatic stellate cells (PSCs), which contributes to pancreatic fibrosis progression [88]. It remains to be explored whether there are ways to modify GLP-1R agonists to enhance their positive effects on pancreatic endocrine and exocrine functions while minimizing their impact on PSCs
In many animal models of other diseases, certain probiotics have been found to induce GLP-1 secretion (Table 2). In addition, using probiotics as oral vectors for recombinant GLP-1R agonist delivery has been explored to replace costly chemical synthesis and inconvenient injections. Probiotics can efficiently target the pancreas, offering a high bioavailability. Lacticaseibacillus paracasei L14 transformed with a plasmid encoding the exendin-4 gene showed efficient secretion and facilitated the transport of exendin-4, enhancing insulin secretion and maintaining β cells [89]. Engineered probiotic yeast Saccharomyces boulardii administered orally also produced bioactive GLP-1R agonists [90]. Apart from delivering GLP-1R agonists, protease-resistant modified GLP-1 (mGLP-1) was constructed with added arginine to ensure the structural integrity of mGLP-1 released in vivo [91]. In addition to producing bioactive GLP-1R agonists, engineered probiotics used as carriers also exert their inherent function of regulating the microbiota. Engineered Clostridium butyricum significantly improved gut microbiota dysbiosis in rats via downregulating the relative abundance of Porphyromonadaceae at the family level and upregulating Lactobacillus at the genus level [92]. Similarly, engineered Escherichia coli Nissle 1917 expressing GLP-1 regulated the intestinal flora and increased the probiotic diversity in mice [93].
An increase in GLP-1 secretion levels has also been observed following the addition of prebiotics, including dietary resistant starch [94,95], resistant maltodextrin [96], fructooligosaccharides [96,97], chondroitin sulfate [98], and Dendrobium officinale polysaccharide (DOP) [99]. The stimulative effect of prebiotics on GLP-1 secretion may occur through stimulating SCFA production [94,95].
Table 2. Summary of probiotics that can promote GLP-1 expression in various disease models.
Table 2. Summary of probiotics that can promote GLP-1 expression in various disease models.
GenusSpeciesDisease ModelsReferences
LacticaseibacillusL. paracasei L-21STC-1 cell line[100]
L. paracasei JY062Glycolipid metabolic disorders[101]
L. casei CCFM419T2DM[102]
L. rhamnosus NCDC 17[103]
LactiplantibacillusL. plantarum subsp. plantarum MTCC5690T2DM[104]
Bifidobacteriumselenium-enriched B. longum DD98T2DM[105]
B. animalis subsp. lactis MN-Gup[106]
B. animalis subsp. lactis NJ241Parkinson’s disease[107]
B. animalis subsp. lactis GCL2505Metabolic syndrome[108]
B. longum subsp. longum B-53STC-1 cell line[100]
AkkermansiaPasteurized A. muciniphilaT2DM[109]
BacteroidesB. thetaiotaomicronalcoholic fatty liver disease[110]
LimosilactobacillusL. fermentum MG4295T2DM[111]
L. fermentum MTCC5689[104]
L. reuteriGlucose metabolism disorder induced by acrylamide; glucose-tolerant humans[112,113]
ClostridiumC. butyricumChronic unpredictable mild stress; T2DM[114,115]
In conclusion, the potential of engineered probiotics to express GLP-1 analogs offers a promising avenue for ameliorating symptoms in CP patients. Despite the current lack of experimental evidence regarding the use of engineered probiotics expressing GLP-1 in CP, their application holds significant promise considering their mechanisms of action and the positive effects observed in other disease models. This approach could improve pancreatic function and manage symptoms more effectively, but further research is needed to fully understand its implications and optimize treatment strategies.

6. Conclusions

Current treatments for CP lack innovation, underscoring the need for novel therapeutic approaches. The gut microbiota can influence pancreatic function through their metabolic activities in the gut, via the gut–pancreas axis. Probiotics and prebiotics may hold potential for treating CP via this axis.
The three possible intervention mechanisms discussed in this review—alleviating small intestine bacterial overgrowth, facilitating SCFAs’ production, and activating GLP-1R in the pancreas—are largely based on theoretical extrapolations from existing research, much of which is derived from other pancreatic disease models. Although there is a scarcity of experimental evidence specifically targeting CP, these mechanisms show strong potential for its treatment, including the improvement of pancreatic endocrine and exocrine functions and maintaining cellular and structural integrity. Therefore, there is an urgent need for experimental validation in the field of chronic pancreatitis. This exploration forms the core focus of this review, highlighting the promising potential of these interventions to address the pressing need for improved chronic pancreatitis therapies.

Author Contributions

Conceptualization, D.W. and Y.P.; methodology, Y.P.; software, Y.P.; validation, Y.P., J.L. and Z.F.; formal analysis, Y.C.; investigation, Y.C.; resources, Z.F.; data curation, Z.F.; writing—original draft preparation, Y.P. and J.L.; writing—review and editing, D.W.; visualization, X.H.; supervision, D.W.; project administration, D.W.; funding acquisition, D.W. and J.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China (grant number 32170788), Beijing Natural Science Foundation (grant number 7232123, 7244390), the National High Level Hospital Clinical Research Funding (grant number 2022-PUMCH-B-023), and the National Key Clinical Specialty Construction Project (grant number ZK108000).

Data Availability Statement

The data underlying this article are available in the article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

CP: chronic pancreatitis; SIBO: small intestinal bacterial overgrowth; SCFA: short-chain fatty acid; GLP-1: glucagon-like peptide-1; GLP-1R: glucagon-like peptide-1 receptor; HDAC: histone deacetylase; CRAMP: cathelicidin-related antimicrobial peptide; NF-κB: nuclear factor kappa-B; GPCR: G-protein-coupled receptor; FFA2: free fatty acid receptor-2; FFA3: free fatty acid receptor-2; T2DM: type 2 diabetes mellitus; cAMP: cyclic adenosine monophosphate; PSC: pancreatic stellate cell.

References

  1. Yadav, D.; Timmons, L.; Benson, J.T.; Dierkhising, R.A.; Chari, S.T. Incidence, prevalence, and survival of chronic pancreatitis: A population-based study. Am. J. Gastroenterol. 2011, 106, 2192–2199. [Google Scholar] [CrossRef] [PubMed]
  2. Yadav, D.; Muddana, V.; O’Connell, M. Hospitalizations for chronic pancreatitis in Allegheny County, Pennsylvania, USA. Pancreatology 2011, 11, 546–552. [Google Scholar] [CrossRef] [PubMed]
  3. Machicado, J.D.; Dudekula, A.; Tang, G.; Xu, H.; Wu, B.U.; Forsmark, C.E.; Yadav, D. Period prevalence of chronic pancreatitis diagnosis from 2001-2013 in the commercially insured population of the United States. Pancreatology 2019, 19, 813–818. [Google Scholar] [CrossRef]
  4. Petrov, M.S.; Olesen, S.S. Metabolic Sequelae: The Pancreatitis Zeitgeist of the 21st Century. Gastroenterology 2023, 165, 1122–1135. [Google Scholar] [CrossRef] [PubMed]
  5. Singh, V.K.; Yadav, D.; Garg, P.K. Diagnosis and Management of Chronic Pancreatitis: A Review. JAMA 2019, 322, 2422–2434. [Google Scholar] [CrossRef]
  6. Thomas, R.M.; Jobin, C. Microbiota in pancreatic health and disease: The next frontier in microbiome research. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 53–64. [Google Scholar] [CrossRef]
  7. Lupu, V.V.; Bratu, R.M.; Trandafir, L.M.; Bozomitu, L.; Paduraru, G.; Gimiga, N.; Ghiga, G.; Forna, L.; Ioniuc, I.; Petrariu, F.D.; et al. Exploring the Microbial Landscape: Gut Dysbiosis and Therapeutic Strategies in Pancreatitis—A Narrative Review. Biomedicines 2024, 12, 645. [Google Scholar] [CrossRef]
  8. Sanders, M.E.; Merenstein, D.J.; Reid, G.; Gibson, G.R.; Rastall, R.A. Author Correction: Probiotics and prebiotics in intestinal health and disease: From biology to the clinic. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 642. [Google Scholar] [CrossRef]
  9. Bures, J.; Cyrany, J.; Kohoutova, D.; Förstl, M.; Rejchrt, S.; Kvetina, J.; Vorisek, V.; Kopacova, M. Small intestinal bacterial overgrowth syndrome. World J. Gastroenterol. 2010, 16, 2978–2990. [Google Scholar] [CrossRef]
  10. Grace, E.; Shaw, C.; Whelan, K.; Andreyev, H.J.N. Review article: Small intestinal bacterial overgrowth-prevalence, clinical features, current and developing diagnostic tests, and treatment. Aliment. Pharmacol. Ther. 2013, 38, 674–688. [Google Scholar] [CrossRef]
  11. Pimentel, M.; Saad, R.J.; Long, M.D.; Rao, S.S.C. ACG Clinical Guideline: Small Intestinal Bacterial Overgrowth. Am. J. Gastroenterol. 2020, 115, 165–178. [Google Scholar] [CrossRef]
  12. Zafar, H.; Jimenez, B.; Schneider, A. Small intestinal bacterial overgrowth: Current update. Curr. Opin. Gastroenterol. 2023, 39, 522–528. [Google Scholar] [CrossRef]
  13. El Kurdi, B.; Babar, S.; El Iskandarani, M.; Bataineh, A.; Lerch, M.M.; Young, M.; Singh, V.P. Factors That Affect Prevalence of Small Intestinal Bacterial Overgrowth in Chronic Pancreatitis: A Systematic Review, Meta-Analysis, and Meta-Regression. Clin. Transl. Gastroenterol. 2019, 10, e00072. [Google Scholar] [CrossRef]
  14. Lee, A.A.; Baker, J.R.; Wamsteker, E.J.; Saad, R.; DiMagno, M.J. Small Intestinal Bacterial Overgrowth Is Common in Chronic Pancreatitis and Associates with Diabetes, Chronic Pancreatitis Severity, Low Zinc Levels, and Opiate Use. Am. J. Gastroenterol. 2019, 114, 1163–1171. [Google Scholar] [CrossRef]
  15. Ní Chonchubhair, H.M.; Bashir, Y.; Dobson, M.; Ryan, B.M.; Duggan, S.N.; Conlon, K.C. The prevalence of small intestinal bacterial overgrowth in non-surgical patients with chronic pancreatitis and pancreatic exocrine insufficiency (PEI). Pancreatology 2018, 18, 379–385. [Google Scholar] [CrossRef]
  16. Capurso, G.; Signoretti, M.; Archibugi, L.; Stigliano, S.; Delle Fave, G. Systematic review and meta-analysis: Small intestinal bacterial overgrowth in chronic pancreatitis. United Eur. Gastroenterol. J. 2016, 4, 697–705. [Google Scholar] [CrossRef]
  17. Rao, S.S.C.; Bhagatwala, J. Small Intestinal Bacterial Overgrowth: Clinical Features and Therapeutic Management. Clin. Transl. Gastroenterol. 2019, 10, e00078. [Google Scholar] [CrossRef]
  18. Shah, S.C.; Day, L.W.; Somsouk, M.; Sewell, J.L. Meta-analysis: Antibiotic therapy for small intestinal bacterial overgrowth. Aliment. Pharmacol. Ther. 2013, 38, 925–934. [Google Scholar] [CrossRef]
  19. Redondo-Cuevas, L.; Belloch, L.; Martín-Carbonell, V.; Nicolás, A.; Alexandra, I.; Sanchis, L.; Ynfante, M.; Colmenares, M.; Mora, M.; Liebana, A.R.; et al. Do Herbal Supplements and Probiotics Complement Antibiotics and Diet in the Management of SIBO? A Randomized Clinical Trial. Nutrients 2024, 16, 1083. [Google Scholar] [CrossRef]
  20. Rosania, R.; Giorgio, F.; Principi, M.; Amoruso, A.; Monno, R.; Di Leo, A.; Ierardi, E. Effect of probiotic or prebiotic supplementation on antibiotic therapy in the small intestinal bacterial overgrowth: A comparative evaluation. Curr. Clin. Pharmacol. 2013, 8, 169–172. [Google Scholar] [CrossRef]
  21. Khalighi, A.R.; Khalighi, M.R.; Behdani, R.; Jamali, J.; Khosravi, A.; Kouhestani, S.; Radmanesh, H.; Esmaeelzadeh, S.; Khalighi, N. Evaluating the efficacy of probiotic on treatment in patients with small intestinal bacterial overgrowth (SIBO)—A pilot study. Indian J. Med. Res. 2014, 140, 604–608. [Google Scholar] [PubMed]
  22. Soifer, L.O.; Peralta, D.; Dima, G.; Besasso, H. Comparative clinical efficacy of a probiotic vs. an antibiotic in the treatment of patients with intestinal bacterial overgrowth and chronic abdominal functional distension: A pilot study. Acta Gastroenterol. Latinoam. 2010, 40, 323–327. [Google Scholar] [PubMed]
  23. Zhong, C.; Qu, C.; Wang, B.; Liang, S.; Zeng, B. Probiotics for Preventing and Treating Small Intestinal Bacterial Overgrowth: A Meta-Analysis and Systematic Review of Current Evidence. J. Clin. Gastroenterol. 2017, 51, 300–311. [Google Scholar] [CrossRef]
  24. Aslan, I.; Tarhan Celebi, L.; Kayhan, H.; Kizilay, E.; Gulbahar, M.Y.; Kurt, H.; Cakici, B. Probiotic Formulations Containing Fixed and Essential Oils Ameliorates SIBO-Induced Gut Dysbiosis in Rats. Pharmaceuticals 2023, 16, 1041. [Google Scholar] [CrossRef]
  25. Barrett, J.S.; Canale, K.E.K.; Gearry, R.B.; Irving, P.M.; Gibson, P.R. Probiotic effects on intestinal fermentation patterns in patients with irritable bowel syndrome. World J. Gastroenterol. 2008, 14, 5020–5024. [Google Scholar] [CrossRef] [PubMed]
  26. Yao, C.K.; Barrett, J.S.; Philpott, H.; Chung, A.R.T.; van Langenberg, D.; Garg, M.; Gibson, P.R. Poor predictive value of breath hydrogen response for probiotic effects in IBS. J. Gastroenterol. Hepatol. 2015, 30, 1731–1739. [Google Scholar] [CrossRef]
  27. Lee, S.-H.; Joo, N.-S.; Kim, K.-M.; Kim, K.-N. The Therapeutic Effect of a Multistrain Probiotic on Diarrhea-Predominant Irritable Bowel Syndrome: A Pilot Study. Gastroenterol. Res. Pract. 2018, 2018, 8791916. [Google Scholar] [CrossRef]
  28. Luo, M.; Liu, Q.; Xiao, L.; Xiong, L.-S. Golden bifid might improve diarrhea-predominant irritable bowel syndrome via microbiota modulation. J. Health Popul. Nutr. 2022, 41, 21. [Google Scholar] [CrossRef]
  29. Bustos Fernández, L.M.; Man, F.; Lasa, J.S. Impact of Saccharomyces boulardii CNCM I-745 on Bacterial Overgrowth and Composition of Intestinal Microbiota in Diarrhea-Predominant Irritable Bowel Syndrome Patients: Results of a Randomized Pilot Study. Dig. Dis. 2023, 41, 798–809. [Google Scholar] [CrossRef]
  30. Hao, Y.; Xu, Y.; Ban, Y.; Li, J.; Wu, B.; Ouyang, Q.; Sun, Z.; Zhang, M.; Cai, Y.; Wang, M.; et al. Efficacy evaluation of probiotics combined with prebiotics in patients with clinical hypothyroidism complicated with small intestinal bacterial overgrowth during the second trimester of pregnancy. Front. Cell Infect. Microbiol. 2022, 12, 983027. [Google Scholar] [CrossRef]
  31. Zhang, M.; Xu, Y.; Sun, Z.; Ban, Y.; Zhai, S.; Wang, W.; Wang, M.; You, J.; Chen, D.; Zhu, S.; et al. Evaluation of probiotics in the treatment of hypothyroidism in early pregnancy combined with small intestinal bacterial overgrowth. Food Sci. Nutr. 2024, 12, 2671–2678. [Google Scholar] [CrossRef]
  32. Ouyang, Q.; Xu, Y.; Ban, Y.; Li, J.; Cai, Y.; Wu, B.; Hao, Y.; Sun, Z.; Zhang, M.; Wang, M.; et al. Probiotics and Prebiotics in Subclinical Hypothyroidism of Pregnancy with Small Intestinal Bacterial Overgrowth. Probiotics Antimicrob. Proteins 2024, 16, 579–588. [Google Scholar] [CrossRef] [PubMed]
  33. García-Collinot, G.; Madrigal-Santillán, E.O.; Martínez-Bencomo, M.A.; Carranza-Muleiro, R.A.; Jara, L.J.; Vera-Lastra, O.; Montes-Cortes, D.H.; Medina, G.; Cruz-Domínguez, M.P. Effectiveness of Saccharomyces boulardii and Metronidazole for Small Intestinal Bacterial Overgrowth in Systemic Sclerosis. Dig. Dis. Sci. 2020, 65, 1134–1143. [Google Scholar] [CrossRef] [PubMed]
  34. Efremova, I.; Maslennikov, R.; Zharkova, M.; Poluektova, E.; Benuni, N.; Kotusov, A.; Demina, T.; Ivleva, A.; Adzhieva, F.; Krylova, T.; et al. Efficacy and Safety of a Probiotic Containing Saccharomyces boulardii CNCM I-745 in the Treatment of Small Intestinal Bacterial Overgrowth in Decompensated Cirrhosis: Randomized, Placebo-Controlled Study. J. Clin. Med. 2024, 13, 919. [Google Scholar] [CrossRef]
  35. Ferolla, S.M.; Couto, C.A.; Costa-Silva, L.; Armiliato, G.N.A.; Pereira, C.A.S.; Martins, F.S.; Ferrari, M.d.L.A.; Vilela, E.G.; Torres, H.O.G.; Cunha, A.S.; et al. Beneficial Effect of Synbiotic Supplementation on Hepatic Steatosis and Anthropometric Parameters, But Not on Gut Permeability in a Population with Nonalcoholic Steatohepatitis. Nutrients 2016, 8, 397. [Google Scholar] [CrossRef] [PubMed]
  36. Kwak, D.S.; Jun, D.W.; Seo, J.G.; Chung, W.S.; Park, S.-E.; Lee, K.N.; Khalid-Saeed, W.; Lee, H.L.; Lee, O.Y.; Yoon, B.C.; et al. Short-term probiotic therapy alleviates small intestinal bacterial overgrowth, but does not improve intestinal permeability in chronic liver disease. Eur. J. Gastroenterol. Hepatol. 2014, 26, 1353–1359. [Google Scholar] [CrossRef]
  37. Liang, S.; Xu, L.; Zhang, D.; Wu, Z. Effect of probiotics on small intestinal bacterial overgrowth in patients with gastric and colorectal cancer. Turk. J. Gastroenterol. 2016, 27, 227–232. [Google Scholar] [CrossRef]
  38. Kanika, G.; Khan, S.; Jena, G. Sodium Butyrate Ameliorates L-Arginine-Induced Pancreatitis and Associated Fibrosis in Wistar Rat: Role of Inflammation and Nitrosative Stress. J. Biochem. Mol. Toxicol. 2015, 29, 349–359. [Google Scholar] [CrossRef]
  39. Pan, L.-L.; Ren, Z.-N.; Yang, J.; Li, B.-B.; Huang, Y.-W.; Song, D.-X.; Li, X.; Xu, J.-J.; Bhatia, M.; Zou, D.-W.; et al. Gut microbiota controls the development of chronic pancreatitis: A critical role of short-chain fatty acids-producing Gram-positive bacteria. Acta Pharm. Sin. B 2023, 13, 4202–4216. [Google Scholar] [CrossRef]
  40. Sun, J.; Furio, L.; Mecheri, R.; van der Does, A.M.; Lundeberg, E.; Saveanu, L.; Chen, Y.; van Endert, P.; Agerberth, B.; Diana, J. Pancreatic β-Cells Limit Autoimmune Diabetes via an Immunoregulatory Antimicrobial Peptide Expressed under the Influence of the Gut Microbiota. Immunity 2015, 43, 304–317. [Google Scholar] [CrossRef]
  41. Vinolo, M.A.R.; Rodrigues, H.G.; Hatanaka, E.; Sato, F.T.; Sampaio, S.C.; Curi, R. Suppressive effect of short-chain fatty acids on production of proinflammatory mediators by neutrophils. J. Nutr. Biochem. 2011, 22, 849–855. [Google Scholar] [CrossRef]
  42. Park, J.-S.; Lee, E.-J.; Lee, J.-C.; Kim, W.-K.; Kim, H.-S. Anti-inflammatory effects of short chain fatty acids in IFN-gamma-stimulated RAW 264.7 murine macrophage cells: Involvement of NF-kappaB and ERK signaling pathways. Int. Immunopharmacol. 2007, 7, 70–77. [Google Scholar] [CrossRef] [PubMed]
  43. Usami, M.; Kishimoto, K.; Ohata, A.; Miyoshi, M.; Aoyama, M.; Fueda, Y.; Kotani, J. Butyrate and trichostatin A attenuate nuclear factor kappaB activation and tumor necrosis factor alpha secretion and increase prostaglandin E2 secretion in human peripheral blood mononuclear cells. Nutr. Res. 2008, 28, 321–328. [Google Scholar] [CrossRef]
  44. Park, G.Y.; Joo, M.; Pedchenko, T.; Blackwell, T.S.; Christman, J.W. Regulation of macrophage cyclooxygenase-2 gene expression by modifications of histone H3. Am. J. Physiol. Lung Cell Mol. Physiol. 2004, 286, L956–L962. [Google Scholar] [CrossRef]
  45. Harada, E.; Kato, S. Effect of short-chain fatty acids on the secretory response of the ovine exocrine pancreas. Am. J. Physiol. 1983, 244, G284–G290. [Google Scholar] [CrossRef] [PubMed]
  46. Katoh, K.; Tsuda, T. Effects of secretagogues on membrane potential and input resistance of pancreatic acinar cells of sheep. Res. Vet. Sci. 1985, 38, 250–251. [Google Scholar] [CrossRef]
  47. Katoh, K.; Tsuda, T. Effects of acetylcholine and short-chain fatty acids on acinar cells of the exocrine pancreas in sheep. J. Physiol. 1984, 356, 479–489. [Google Scholar] [CrossRef]
  48. Katoh, K.; Tsuda, T. Effects of intravenous injection of butyrate on the exocrine pancreatic secretion in guinea pigs. Comp. Biochem. Physiol. A Comp. Physiol. 1987, 87, 569–572. [Google Scholar] [CrossRef]
  49. Lu, V.B.; Gribble, F.M.; Reimann, F. Free Fatty Acid Receptors in Enteroendocrine Cells. Endocrinology 2018, 159, 2826–2835. [Google Scholar] [CrossRef]
  50. Teyani, R.; Moniri, N.H. Gut feelings in the islets: The role of the gut microbiome and the FFA2 and FFA3 receptors for short chain fatty acids on β-cell function and metabolic regulation. Br. J. Pharmacol. 2023, 180, 3113–3129. [Google Scholar] [CrossRef]
  51. Rosli, N.S.A.; Abd Gani, S.; Khayat, M.E.; Zaidan, U.H.; Ismail, A.; Abdul Rahim, M.B.H. Short-chain fatty acids: Possible regulators of insulin secretion. Mol. Cell Biochem. 2023, 478, 517–530. [Google Scholar] [CrossRef]
  52. Frost, F.; Weiss, F.U.; Sendler, M.; Kacprowski, T.; Rühlemann, M.; Bang, C.; Franke, A.; Völker, U.; Völzke, H.; Lamprecht, G.; et al. The Gut Microbiome in Patients with Chronic Pancreatitis Is Characterized by Significant Dysbiosis and Overgrowth by Opportunistic Pathogens. Clin. Transl. Gastroenterol. 2020, 11, e00232. [Google Scholar] [CrossRef]
  53. Jandhyala, S.M.; Madhulika, A.; Deepika, G.; Rao, G.V.; Reddy, D.N.; Subramanyam, C.; Sasikala, M.; Talukdar, R. Altered intestinal microbiota in patients with chronic pancreatitis: Implications in diabetes and metabolic abnormalities. Sci. Rep. 2017, 7, 43640. [Google Scholar] [CrossRef]
  54. Moens, F.; Van den Abbeele, P.; Basit, A.W.; Dodoo, C.; Chatterjee, R.; Smith, B.; Gaisford, S. A four-strain probiotic exerts positive immunomodulatory effects by enhancing colonic butyrate production in vitro. Int. J. Pharm. 2019, 555, 1–10. [Google Scholar] [CrossRef]
  55. Yang, S.; Shang, J.; Liu, L.; Tang, Z.; Meng, X. Strains producing different short-chain fatty acids alleviate DSS-induced ulcerative colitis by regulating intestinal microecology. Food Funct. 2022, 13, 12156–12169. [Google Scholar] [CrossRef]
  56. Zhang, Y.; Li, Y.; Wang, X.; Huang, J.; Feng, X.; Shi, C.; Yang, W.; Jiang, Y.; Cao, X.; Wang, J.; et al. Lactobacillus Plantarum NC8 and its metabolite acetate alleviate type 1 diabetes via inhibiting NLRP3. Microb. Pathog. 2023, 182, 106237. [Google Scholar] [CrossRef]
  57. Miao, X.; Jiang, Y.; Kong, D.; Wu, Z.; Liu, H.; Ye, X.; Gong, W. Lactobacillus rhamnosus HN001 Ameliorates BEZ235-Induced Intestinal Dysbiosis and Prolongs Cardiac Transplant Survival. Microbiol. Spectr. 2022, 10, e0079422. [Google Scholar] [CrossRef]
  58. Xu, B.; Wang, Z.; Wang, Y.; Zhang, K.; Li, J.; Zhou, L.; Li, B. Milk-derived Lactobacillus with high production of short-chain fatty acids relieves antibiotic-induced diarrhea in mice. Food Funct. 2024, 15, 5329–5342. [Google Scholar] [CrossRef]
  59. Byrne, C.S.; Chambers, E.S.; Morrison, D.J.; Frost, G. The role of short chain fatty acids in appetite regulation and energy homeostasis. Int. J. Obes. 2015, 39, 1331–1338. [Google Scholar] [CrossRef]
  60. Morrison, D.J.; Preston, T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes 2016, 7, 189–200. [Google Scholar] [CrossRef]
  61. Pascale, N.; Gu, F.; Larsen, N.; Jespersen, L.; Respondek, F. The Potential of Pectins to Modulate the Human Gut Microbiota Evaluated by In Vitro Fermentation: A Systematic Review. Nutrients 2022, 14, 3629. [Google Scholar] [CrossRef]
  62. Macfarlane, S.; Macfarlane, G.T. Regulation of short-chain fatty acid production. Proc. Nutr. Soc. 2003, 62, 67–72. [Google Scholar] [CrossRef]
  63. Boets, E.; Deroover, L.; Houben, E.; Vermeulen, K.; Gomand, S.V.; Delcour, J.A.; Verbeke, K. Quantification of In Vivo Colonic Short Chain Fatty Acid Production from Inulin. Nutrients 2015, 7, 8916–8929. [Google Scholar] [CrossRef]
  64. François, I.E.J.A.; Lescroart, O.; Veraverbeke, W.S.; Marzorati, M.; Possemiers, S.; Evenepoel, P.; Hamer, H.; Houben, E.; Windey, K.; Welling, G.W.; et al. Effects of a wheat bran extract containing arabinoxylan oligosaccharides on gastrointestinal health parameters in healthy adult human volunteers: A double-blind, randomised, placebo-controlled, cross-over trial. Br. J. Nutr. 2012, 108, 2229–2242. [Google Scholar] [CrossRef]
  65. Guimarães, J.B.; Rodrigues, V.F.; Pereira, Í.S.; Manso, G.M.d.C.; Elias-Oliveira, J.; Leite, J.A.; Waldetario, M.C.G.M.; de Oliveira, S.; Gomes, A.B.D.S.P.; Faria, A.M.C.; et al. Inulin prebiotic ameliorates type 1 diabetes dictating regulatory T cell homing via CCR4 to pancreatic islets and butyrogenic gut microbiota in murine model. J. Leukoc. Biol. 2024, 115, 483–496. [Google Scholar] [CrossRef]
  66. Falony, G.; Vlachou, A.; Verbrugghe, K.; De Vuyst, L. Cross-feeding between Bifidobacterium longum BB536 and acetate-converting, butyrate-producing colon bacteria during growth on oligofructose. Appl. Environ. Microbiol. 2006, 72, 7835–7841. [Google Scholar] [CrossRef]
  67. Moens, F.; Weckx, S.; De Vuyst, L. Bifidobacterial inulin-type fructan degradation capacity determines cross-feeding interactions between bifidobacteria and Faecalibacterium prausnitzii. Int. J. Food Microbiol. 2016, 231, 76–85. [Google Scholar] [CrossRef]
  68. Drucker, D.J. Mechanisms of Action and Therapeutic Application of Glucagon-like Peptide-1. Cell Metab. 2018, 27, 740–756. [Google Scholar] [CrossRef]
  69. Müller, T.D.; Finan, B.; Bloom, S.R.; D’Alessio, D.; Drucker, D.J.; Flatt, P.R.; Fritsche, A.; Gribble, F.; Grill, H.J.; Habener, J.F.; et al. Glucagon-like peptide 1 (GLP-1). Mol. Metab. 2019, 30, 72–130. [Google Scholar] [CrossRef]
  70. Nogueiras, R.; Nauck, M.A.; Tschöp, M.H. Gut hormone co-agonists for the treatment of obesity: From bench to bedside. Nat. Metab. 2023, 5, 933–944. [Google Scholar] [CrossRef]
  71. Drucker, D.J.; Habener, J.F.; Holst, J.J. Discovery, characterization, and clinical development of the glucagon-like peptides. J. Clin. Investig. 2017, 127, 4217–4227. [Google Scholar] [CrossRef]
  72. Richards, P.; Parker, H.E.; Adriaenssens, A.E.; Hodgson, J.M.; Cork, S.C.; Trapp, S.; Gribble, F.M.; Reimann, F. Identification and characterization of GLP-1 receptor-expressing cells using a new transgenic mouse model. Diabetes 2014, 63, 1224–1233. [Google Scholar] [CrossRef]
  73. Waser, B.; Blank, A.; Karamitopoulou, E.; Perren, A.; Reubi, J.C. Glucagon-like-peptide-1 receptor expression in normal and diseased human thyroid and pancreas. Mod. Pathol. 2015, 28, 391–402. [Google Scholar] [CrossRef]
  74. de Heer, J.; Rasmussen, C.; Coy, D.H.; Holst, J.J. Glucagon-like peptide-1, but not glucose-dependent insulinotropic peptide, inhibits glucagon secretion via somatostatin (receptor subtype 2) in the perfused rat pancreas. Diabetologia 2008, 51, 2263–2270. [Google Scholar] [CrossRef]
  75. Campbell, J.E.; Drucker, D.J. Pharmacology, physiology, and mechanisms of incretin hormone action. Cell Metab. 2013, 17, 819–837. [Google Scholar] [CrossRef]
  76. Yusta, B.; Baggio, L.L.; Estall, J.L.; Koehler, J.A.; Holland, D.P.; Li, H.; Pipeleers, D.; Ling, Z.; Drucker, D.J. GLP-1 receptor activation improves beta cell function and survival following induction of endoplasmic reticulum stress. Cell Metab. 2006, 4, 391–406. [Google Scholar] [CrossRef]
  77. Lamont, B.J.; Andrikopoulos, S. Hope and fear for new classes of type 2 diabetes drugs: Is there preclinical evidence that incretin-based therapies alter pancreatic morphology? J. Endocrinol. 2014, 221, T43–T61. [Google Scholar] [CrossRef] [PubMed]
  78. Rankin, M.M.; Kushner, J.A. Adaptive beta-cell proliferation is severely restricted with advanced age. Diabetes 2009, 58, 1365–1372. [Google Scholar] [CrossRef]
  79. Tschen, S.-I.; Georgia, S.; Dhawan, S.; Bhushan, A. Skp2 is required for incretin hormone-mediated β-cell proliferation. Mol. Endocrinol. 2011, 25, 2134–2143. [Google Scholar] [CrossRef] [PubMed]
  80. Fiorentino, T.V.; Owston, M.; Abrahamian, G.; La Rosa, S.; Marando, A.; Perego, C.; Di Cairano, E.S.; Finzi, G.; Capella, C.; Sessa, F.; et al. Chronic continuous exenatide infusion does not cause pancreatic inflammation and ductal hyperplasia in non-human primates. Am. J. Pathol. 2015, 185, 139–150. [Google Scholar] [CrossRef]
  81. Hou, Y.; Ernst, S.A.; Heidenreich, K.; Williams, J.A. Glucagon-like peptide-1 receptor is present in pancreatic acinar cells and regulates amylase secretion through cAMP. Am. J. Physiol. Gastrointest. Liver Physiol. 2016, 310, G26–G33. [Google Scholar] [CrossRef]
  82. Pyke, C.; Heller, R.S.; Kirk, R.K.; Ørskov, C.; Reedtz-Runge, S.; Kaastrup, P.; Hvelplund, A.; Bardram, L.; Calatayud, D.; Knudsen, L.B. GLP-1 receptor localization in monkey and human tissue: Novel distribution revealed with extensively validated monoclonal antibody. Endocrinology 2014, 155, 1280–1290. [Google Scholar] [CrossRef]
  83. Kirk, R.K.; Pyke, C.; von Herrath, M.G.; Hasselby, J.P.; Pedersen, L.; Mortensen, P.G.; Knudsen, L.B.; Coppieters, K. Immunohistochemical assessment of glucagon-like peptide 1 receptor (GLP-1R) expression in the pancreas of patients with type 2 diabetes. Diabetes Obes. Metab. 2017, 19, 705–712. [Google Scholar] [CrossRef]
  84. Koehler, J.A.; Baggio, L.L.; Lamont, B.J.; Ali, S.; Drucker, D.J. Glucagon-like peptide-1 receptor activation modulates pancreatitis-associated gene expression but does not modify the susceptibility to experimental pancreatitis in mice. Diabetes 2009, 58, 2148–2161. [Google Scholar] [CrossRef]
  85. Koehler, J.A.; Baggio, L.L.; Cao, X.; Abdulla, T.; Campbell, J.E.; Secher, T.; Jelsing, J.; Larsen, B.; Drucker, D.J. Glucagon-like peptide-1 receptor agonists increase pancreatic mass by induction of protein synthesis. Diabetes 2015, 64, 1046–1056. [Google Scholar] [CrossRef]
  86. Steinberg, W.M.; Rosenstock, J.; Wadden, T.A.; Donsmark, M.; Jensen, C.B.; DeVries, J.H. Impact of Liraglutide on Amylase, Lipase, and Acute Pancreatitis in Participants with Overweight/Obesity and Normoglycemia, Prediabetes, or Type 2 Diabetes: Secondary Analyses of Pooled Data from the SCALE Clinical Development Program. Diabetes Care 2017, 40, 839–848, Erratum in Diabetes Care 2018, 41, 1538. [Google Scholar] [CrossRef]
  87. Knapen, L.M.; de Jong, R.G.P.J.; Driessen, J.H.M.; Keulemans, Y.C.; van Erp, N.P.; De Bruin, M.L.; Leufkens, H.G.M.; Croes, S.; de Vries, F. Use of incretin agents and risk of acute and chronic pancreatitis: A population-based cohort study. Diabetes Obes. Metab. 2017, 19, 401–411. [Google Scholar] [CrossRef]
  88. Yang, Y.; Yu, X.; Huang, L.; Yu, C. GLP-1R agonist may activate pancreatic stellate cells to induce rat pancreatic tissue lesion. Pancreatology 2013, 13, 498–501. [Google Scholar] [CrossRef]
  89. Zeng, Z.; Yu, R.; Zuo, F.; Zhang, B.; Peng, D.; Ma, H.; Chen, S. Heterologous Expression and Delivery of Biologically Active Exendin-4 by Lactobacillus paracasei L14. PLoS ONE 2016, 11, e0165130. [Google Scholar] [CrossRef]
  90. Hedin, K.A.; Zhang, H.; Kruse, V.; Rees, V.E.; Bäckhed, F.; Greiner, T.U.; Vazquez-Uribe, R.; Sommer, M.O.A. Cold Exposure and Oral Delivery of GLP-1R Agonists by an Engineered Probiotic Yeast Strain Have Antiobesity Effects in Mice. ACS Synth. Biol. 2023, 12, 3433–3442. [Google Scholar] [CrossRef]
  91. Wang, Q.; Guo, H.; Mao, W.; Qian, X.; Liu, Y. The Oral Delivery System of Modified GLP-1 by Probiotics for T2DM. Pharmaceutics 2023, 15, 1202. [Google Scholar] [CrossRef]
  92. Wang, X.-L.; Chen, W.-J.; Jin, R.; Xu, X.; Wei, J.; Huang, H.; Tang, Y.-H.; Zou, C.-W.; Chen, T.-T. Engineered probiotics Clostridium butyricum-pMTL007-GLP-1 improves blood pressure via producing GLP-1 and modulating gut microbiota in spontaneous hypertension rat models. Microb. Biotechnol. 2023, 16, 799–812. [Google Scholar] [CrossRef]
  93. Wang, Y.; Shi, Y.; Peng, X.; Li, T.; Liang, C.; Wang, W.; Zhou, M.; Yang, J.; Cheng, J.; Zhang, Z.; et al. Biochemotaxis-Oriented Engineering Bacteria Expressing GLP-1 Enhance Diabetes Therapy by Regulating the Balance of Immune. Adv. Healthc. Mater. 2024, 13, e2303958. [Google Scholar] [CrossRef]
  94. Zhou, J.; Martin, R.J.; Tulley, R.T.; Raggio, A.M.; McCutcheon, K.L.; Shen, L.; Danna, S.C.; Tripathy, S.; Hegsted, M.; Keenan, M.J. Dietary resistant starch upregulates total GLP-1 and PYY in a sustained day-long manner through fermentation in rodents. Am. J. Physiol. Endocrinol. Metab. 2008, 295, E1160–E1166. [Google Scholar] [CrossRef]
  95. Shen, L.; Keenan, M.J.; Raggio, A.; Williams, C.; Martin, R.J. Dietary-resistant starch improves maternal glycemic control in Goto-Kakizaki rat. Mol. Nutr. Food Res. 2011, 55, 1499–1508. [Google Scholar] [CrossRef]
  96. Hira, T.; Suto, R.; Kishimoto, Y.; Kanahori, S.; Hara, H. Resistant maltodextrin or fructooligosaccharides promotes GLP-1 production in male rats fed a high-fat and high-sucrose diet, and partially reduces energy intake and adiposity. Eur. J. Nutr. 2018, 57, 965–979. [Google Scholar] [CrossRef]
  97. Wongkrasant, P.; Pongkorpsakol, P.; Chitwattananont, S.; Satianrapapong, W.; Tuangkijkul, N.; Muanprasat, C. Fructo-oligosaccharides alleviate inflammation-associated apoptosis of GLP-1 secreting L cells via inhibition of iNOS and cleaved caspase-3 expression. J. Pharmacol. Sci. 2020, 143, 65–73. [Google Scholar] [CrossRef]
  98. Pichette, J.; Fynn-Sackey, N.; Gagnon, J. Hydrogen Sulfide and Sulfate Prebiotic Stimulates the Secretion of GLP-1 and Improves Glycemia in Male Mice. Endocrinology 2017, 158, 3416–3425. [Google Scholar] [CrossRef]
  99. Liu, H.; Xing, Y.; Wang, Y.; Ren, X.; Zhang, D.; Dai, J.; Xiu, Z.; Yu, S.; Dong, Y. Dendrobium officinale Polysaccharide Prevents Diabetes via the Regulation of Gut Microbiota in Prediabetic Mice. Foods 2023, 12, 2310. [Google Scholar] [CrossRef]
  100. Cheng, Z.; Chen, J.; Zhang, Y.; Li, X.; Zhang, N.; Liu, F.; Jiao, Y. In Vitro Hypoglycemic Activities of Lactobacilli and Bifidobacterium Strains from Healthy Children’s Sources and Their Effect on Stimulating GLP-1 Secretion in STC-1 Cells. Foods 2024, 13, 519. [Google Scholar] [CrossRef]
  101. Su, Y.; Ren, J.; Zhang, J.; Zheng, J.; Zhang, Q.; Tian, Y.; Zhang, Y.; Jiang, Y.; Zhang, W. Lactobacillus paracasei JY062 Alleviates Glucolipid Metabolism Disorders via the Adipoinsular Axis and Gut Microbiota. Nutrients 2024, 16, 267. [Google Scholar] [CrossRef] [PubMed]
  102. Wang, G.; Li, X.; Zhao, J.; Zhang, H.; Chen, W. Lactobacillus casei CCFM419 attenuates type 2 diabetes via a gut microbiota dependent mechanism. Food Funct. 2017, 8, 3155–3164. [Google Scholar] [CrossRef]
  103. Singh, S.; Sharma, R.K.; Malhotra, S.; Pothuraju, R.; Shandilya, U.K. Lactobacillus rhamnosus NCDC17 ameliorates type-2 diabetes by improving gut function, oxidative stress and inflammation in high-fat-diet fed and streptozotocintreated rats. Benef. Microbes 2017, 8, 243–255. [Google Scholar] [CrossRef]
  104. Balakumar, M.; Prabhu, D.; Sathishkumar, C.; Prabu, P.; Rokana, N.; Kumar, R.; Raghavan, S.; Soundarajan, A.; Grover, S.; Batish, V.K.; et al. Improvement in glucose tolerance and insulin sensitivity by probiotic strains of Indian gut origin in high-fat diet-fed C57BL/6J mice. Eur. J. Nutr. 2018, 57, 279–295. [Google Scholar] [CrossRef]
  105. Zhao, D.; Zhu, H.; Gao, F.; Qian, Z.; Mao, W.; Yin, Y.; Tan, J.; Chen, D. Antidiabetic effects of selenium-enriched Bifidobacterium longum DD98 in type 2 diabetes model of mice. Food Funct. 2020, 11, 6528–6541. [Google Scholar] [CrossRef]
  106. Zhang, C.; Fang, B.; Zhang, N.; Zhang, Q.; Niu, T.; Zhao, L.; Sun, E.; Wang, J.; Xiao, R.; He, J.; et al. The Effect of Bifidobacterium animalis subsp. lactis MN-Gup on Glucose Metabolism, Gut Microbiota, and Their Metabolites in Type 2 Diabetic Mice. Nutrients 2024, 16, 1691. [Google Scholar] [CrossRef]
  107. Dong, Y.; Qi, Y.; Chen, J.; Han, S.; Su, W.; Ma, X.; Yu, Y.; Wang, Y. Neuroprotective Effects of Bifidobacterium animalis subsp. lactis NJ241 in a Mouse Model of Parkinson’s Disease: Implications for Gut Microbiota and PGC-1α. Mol. Neurobiol. 2024, 1–15. [Google Scholar] [CrossRef]
  108. Aoki, R.; Kamikado, K.; Suda, W.; Takii, H.; Mikami, Y.; Suganuma, N.; Hattori, M.; Koga, Y. A proliferative probiotic Bifidobacterium strain in the gut ameliorates progression of metabolic disorders via microbiota modulation and acetate elevation. Sci. Rep. 2017, 7, 43522. [Google Scholar] [CrossRef] [PubMed]
  109. Niu, H.; Zhou, M.; Ji, A.; Zogona, D.; Wu, T.; Xu, X. Molecular Mechanism of Pasteurized Akkermansia muciniphila in Alleviating Type 2 Diabetes Symptoms. J. Agric. Food Chem. 2024, 72, 13083–13098. [Google Scholar] [CrossRef] [PubMed]
  110. Sangineto, M.; Grander, C.; Grabherr, F.; Mayr, L.; Enrich, B.; Schwärzler, J.; Dallio, M.; Bukke, V.N.; Moola, A.; Moschetta, A.; et al. Recovery of Bacteroides thetaiotaomicron ameliorates hepatic steatosis in experimental alcohol-related liver disease. Gut Microbes 2022, 14, 2089006. [Google Scholar] [CrossRef]
  111. Kim, J.E.; Lee, J.Y.; Kang, C.-H. Limosilactobacillus fermentum MG4295 Improves Hyperglycemia in High-Fat Diet-Induced Mice. Foods 2022, 11, 231. [Google Scholar] [CrossRef]
  112. Yue, Z.; Zhao, F.; Guo, Y.; Zhang, Y.; Chen, Y.; He, L.; Li, L. Lactobacillus reuteri JCM 1112 ameliorates chronic acrylamide-induced glucose metabolism disorder via the bile acid-TGR5-GLP-1 axis and modulates intestinal oxidative stress in mice. Food Funct. 2024, 15, 6450–6458. [Google Scholar] [CrossRef]
  113. Simon, M.-C.; Strassburger, K.; Nowotny, B.; Kolb, H.; Nowotny, P.; Burkart, V.; Zivehe, F.; Hwang, J.-H.; Stehle, P.; Pacini, G.; et al. Intake of Lactobacillus reuteri improves incretin and insulin secretion in glucose-tolerant humans: A proof of concept. Diabetes Care 2015, 38, 1827–1834. [Google Scholar] [CrossRef]
  114. Sun, J.; Wang, F.; Hu, X.; Yang, C.; Xu, H.; Yao, Y.; Liu, J. Clostridium butyricum Attenuates Chronic Unpredictable Mild Stress-Induced Depressive-Like Behavior in Mice via the Gut-Brain Axis. J. Agric. Food Chem. 2018, 66, 8415–8421. [Google Scholar] [CrossRef]
  115. Jia, L.; Li, D.; Feng, N.; Shamoon, M.; Sun, Z.; Ding, L.; Zhang, H.; Chen, W.; Sun, J.; Chen, Y.Q. Anti-diabetic Effects of Clostridium butyricum CGMCC0313.1 through Promoting the Growth of Gut Butyrate-producing Bacteria in Type 2 Diabetic Mice. Sci. Rep. 2017, 7, 7046. [Google Scholar] [CrossRef]
Figure 1. Bidirectional gut–pancreas interactions in the context of CP. The arrow indicates that one side has influence over the other. SCFA, short-chain fatty acid; GLP-1, glucagon-like peptide 1; SIBO, small intestinal bacterial overgrowth; and AMP, antimicrobial peptide. This figure was created with BioRender.com (accessed in July 2024).
Figure 1. Bidirectional gut–pancreas interactions in the context of CP. The arrow indicates that one side has influence over the other. SCFA, short-chain fatty acid; GLP-1, glucagon-like peptide 1; SIBO, small intestinal bacterial overgrowth; and AMP, antimicrobial peptide. This figure was created with BioRender.com (accessed in July 2024).
Microorganisms 12 01760 g001
Figure 2. Flow chart of literature search for this review. Asterisks represent 0 to multiple characters (PubMed search tips).
Figure 2. Flow chart of literature search for this review. Asterisks represent 0 to multiple characters (PubMed search tips).
Microorganisms 12 01760 g002
Table 1. Main examples of probiotics producing short-chain fatty acids.
Table 1. Main examples of probiotics producing short-chain fatty acids.
ProbioticsProductsReferences
Bifidobacterium bifidum H3-R2Acetate[55]
Lactiplantibacillus plantarum NC8Acetate[56]
Lacticaseibacillus rhamnosus HN001Propionate[57]
Propionibacterium freudenreichii B1Propionate[55]
Clostridium butyricum C1–6Butyrate[55]
Lactobacillus acidophilus KLDS 1.0901Acetate, propionate, butyrate[58]
Lactiplantibacillus plantarum KLDS 1.0386
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pan, Y.; Li, J.; Fan, Z.; Chen, Y.; Huang, X.; Wu, D. New Insights into Chronic Pancreatitis: Potential Mechanisms Related to Probiotics. Microorganisms 2024, 12, 1760. https://doi.org/10.3390/microorganisms12091760

AMA Style

Pan Y, Li J, Fan Z, Chen Y, Huang X, Wu D. New Insights into Chronic Pancreatitis: Potential Mechanisms Related to Probiotics. Microorganisms. 2024; 12(9):1760. https://doi.org/10.3390/microorganisms12091760

Chicago/Turabian Style

Pan, Yingyu, Jianing Li, Zhengyang Fan, Yonghao Chen, Xiaoxuan Huang, and Dong Wu. 2024. "New Insights into Chronic Pancreatitis: Potential Mechanisms Related to Probiotics" Microorganisms 12, no. 9: 1760. https://doi.org/10.3390/microorganisms12091760

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

Article Metrics

Back to TopTop