1
|
Nikola L, Iva L. Gut microbiota as a modulator of type 1 diabetes: A molecular perspective. Life Sci 2024; 359:123187. [PMID: 39488260 DOI: 10.1016/j.lfs.2024.123187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 10/04/2024] [Accepted: 10/24/2024] [Indexed: 11/04/2024]
Abstract
Type 1 diabetes (T1D) is defined as an autoimmune metabolic disorder, characterized by destruction of pancreatic β-cells and high blood sugar levels. If left untreated, T1D results in severe health complications, including cardiovascular and kidney disease, as well as nerve damage, with ultimately grave consequences. Besides the role of genetic and certain environmental factors in T1D development, in the last decade, one new player emerged to affect T1D pathology as well, and that is a gut microbiota. Dysbiosis of gut bacteria can contribute to T1D by gut barrier disruption and the activation of autoimmune response, leading to the destruction of insulin producing cells, causing the development and aggravation of T1D symptoms. The relationship between gut microbiota and diabetes is complex and varies between individuals and additional research is needed to fully understand the effects of gut microbiome alternations in T1D pathogenesis. Therefore, the goal of this review is to understand the current knowledge in underlying molecular mechanism of gut microbiota effects, which leads to the new approaches for further studies in the prevention and treatment of T1D.
Collapse
Affiliation(s)
- Lukic Nikola
- Laboratory for Molecular Biology and Endocrinology, Institute of Nuclear Sciences "Vinca", National Institute of the Republic of Serbia, University of Belgrade, Serbia
| | - Lukic Iva
- Laboratory for Molecular Biology and Endocrinology, Institute of Nuclear Sciences "Vinca", National Institute of the Republic of Serbia, University of Belgrade, Serbia.
| |
Collapse
|
2
|
Ruathong W, Khuituan P, Peerakietkhajorn S, Teanpaisan R, Nopparat J. The probiotic Lacticaseibacillus rhamnosus SD11 alleviates the progression of liver and colon damage through modulation of inflammation and tight junction proteins in streptozotocin-induced diabetic mice. PLoS One 2024; 19:e0313395. [PMID: 39570868 PMCID: PMC11581286 DOI: 10.1371/journal.pone.0313395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024] Open
Abstract
Lacticaseibacillus rhamnosus SD11 (SD11) has several health benefits for the host, including antidiabetic, anti-inflammatory, and antimicrobial effects. However, the antidiabetic mechanism of SD11 has not been clearly elucidated. The current study assessed the effects of SD11 and the associated underlying mechanisms on streptozotocin (STZ)-induced diabetic mice. Compared with the normal control, SD11 supplementation for 4 weeks significantly improved the metabolic profiles, including body weight (BW), fasting blood glucose (FBG), fasting insulin level (FIN), and liver index (LI), in conjunction with a lower NAS score. A notable reduction in the liver function parameters aminotransferase (AST), alanine aminotransferase (ALT), and alkaline phosphatase (ALP) and total cholesterol (TC), together with histopathology studies, supported diabetic recovery by SD11. A closer examination of two major markers for the insulin pathway, insulin receptor (INSR) and insulin substrate (IRS)-1, revealed that SD11 could exert its glucose control through the upregulation of these molecules, which were almost demolished in nontreated diabetic livers. Additionally, SD11-treated mice exhibited alleviation of oxidative stress enzymes; downregulation of proinflammatory cytokines, including interleukin (IL)-6, IL-1β, tumor necrosis factor (TNF)-α, and interferon (IFN)-γ; and decreased infiltration of macrophages into liver tissue. These findings were concomitant with the preservation of the tight junction proteins occludin and zona occludin (ZO)-1, which in turn lowered the levels of the inflammatory cytokines IL-1β and TNF-α and prevented colon tissue injury to some extent. Notably, the results for the SD11 control mice were identical to those for the normal control mice. Overall, our findings that SD11 delays liver deterioration and reduces colon lesions in diabetic mice provide evidence for the use of SD11 as an effective strategy to improve diabetes-related symptoms.
Collapse
Affiliation(s)
- Waraporn Ruathong
- Faculty of Science, Division of Health and Applied Sciences, Prince of Songkla University, Songkhla, Thailand
| | - Pissared Khuituan
- Faculty of Science, Division of Health and Applied Sciences, Prince of Songkla University, Songkhla, Thailand
| | - Saranya Peerakietkhajorn
- Faculty of Science, Division of Biological Science, Prince of Songkla University, Songkhla, Thailand
| | - Rawee Teanpaisan
- Faculty of Dentistry, Research Center of Excellence for Oral Health, Prince of Songkla University, Hat Yai, Thailand
| | - Jongdee Nopparat
- Faculty of Science, Division of Health and Applied Sciences, Prince of Songkla University, Songkhla, Thailand
- Center of Excellence for Trace Analysis and Biosensor, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| |
Collapse
|
3
|
Su Z, Bian L, Zhao H, Yang C, Gu Y, Cai Y, Yang T, Xu X. KIR2DL5 +CD8 + T cells associate with dietary lipid intake and are active in type 1 diabetes. Int Immunopharmacol 2024; 141:112971. [PMID: 39178517 DOI: 10.1016/j.intimp.2024.112971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUND Recent studies have shown that KIR+CD8+ T cells play a role in suppressing autoimmunity by eliminating pathogenic CD4+ T cells. However, their specific role in type 1 diabetes (T1D) remains unclear. METHODS In this study, we enrolled 108 patients diagnosed with T1D and 86 healthy individuals. We conducted flow cytometric analysis to examine the various subtypes of KIR+CD8+ T cells derived from peripheral blood mononuclear cells. Additionally, CD8+ T cells were isolated from the peripheral blood of T1D patients to assess the functions of different KIR+CD8+ T cell subtypes. To investigate the influence of lipids on the characteristics and activities of these T cell subtypes, the isolated CD8+ T cells were cultured with varying concentrations of palmitic acid (PA). Furthermore, we utilized an NSG (NOD scid gamma) mouse adoptive transfer model to assess the impact of dietary lipid intake on the functionality of KIR2DL5+CD8+ T cells in vivo. RESULTS We observed variations in circulating KIR+CD8+ T cell subtypes between patients with T1D and healthy controls. Notably, we observed a significant negative correlation between the frequencies of circulating KIR+CD8+ T cells and the titers of ZnT8 autoantibodies in individuals with T1D. Among these subtypes, KIR2DL5+CD8+ T cells demonstrated a positive association with dietary fat intake, characterized by increased perforin expression and reduced PD-1 expression. Importantly, KIR2DL5+CD8+ T cells exhibited enhanced proliferative capacity compared to other KIR+CD8+ T cell subsets. Palmitic acid (PA) was found to enhance the activation of KIR2DL5+CD8+ T cells and strengthened their ability to suppress CD4+ T cell proliferation in T1D patients. Moreover, dietary lipid intake significantly enhanced the functionality of KIR2DL5+CD8+ T cells in an NSG mouse adoptive transfer model. CONCLUSION Our findings suggest that lipid intake enhances the functionality of human KIR2DL5+CD8+ T cells and may offer implications for immunotherapy in T1D.
Collapse
Affiliation(s)
- Zhangyao Su
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Lingling Bian
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China; Department of Endocrinology, The First People's Hospital of Yancheng, Yancheng, Jiangsu Province, China
| | - Hang Zhao
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Chun Yang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yong Gu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yun Cai
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Tao Yang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Xinyu Xu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
4
|
Jonić N, Koprivica I, Kyrkou SG, Bistas VP, Chatzigiannis C, Radulović N, Pilipović I, Jovanović A, Jovanović MB, Dimitrijević M, Tzakos AG, Stojanović I. Novel AHR ligand AGT-5 ameliorates type 1 diabetes in mice through regulatory cell activation in the early phase of the disease. Front Immunol 2024; 15:1454156. [PMID: 39308860 PMCID: PMC11412818 DOI: 10.3389/fimmu.2024.1454156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 08/23/2024] [Indexed: 09/25/2024] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease with a strong chronic inflammatory component. One possible strategy for the treatment of T1D is to stimulate the regulatory arm of the immune response, i.e. to promote the function of tolerogenic dendritic cells (tolDC) and regulatory T cells (Treg). Since both cell types have been shown to be responsive to the aryl hydrocarbon receptor (AHR) activation, we used a recently characterized member of a new class of fluorescent AHR ligands, AGT-5, to modulate streptozotocin-induced T1D in C57BL/6 mice. Prophylactic oral administration of AGT-5 reduced hyperglycemia and insulitis in these mice. Phenotypic and functional analysis of cells in the pancreatic infiltrates of AGT-5-treated mice (at the early phase of T1D) revealed a predominantly anti-inflammatory environment, as evidenced by the upregulation of tolDC and Treg frequency, while CD8+ cell, Th1 and Th17 cells were significantly reduced. Similarly, AGT-5 enhanced the proportion of Treg and tolDC in small intestine lamina propria and suppressed the activation status of antigen-presenting cells through down-regulation of co-stimulatory molecules CD40, CD80 and CD86. The expression levels of Cyp1a1, controlled by the AHR, were increased in CD4+, CD8+ and Treg, confirming the AHR-mediated effect of AGT-5 in these cells. Finally, AGT-5 stimulated the function of regulatory cells in the pancreatic islets and lamina propria by upregulating indoleamine 2,3-dioxigenase 1 (IDO1) in tolDC. These findings were supported by the abrogation of AGT-5-mediated in vitro effects on DC in the presence of IDO1 inhibitor. AGT-5 also increased the expression of CD39 or CD73 ATP-degrading ectoenzymes by Treg. The increase in Treg is further supported by the upregulated frequency of IL-2-producing type 3 innate lymphoid cells (ILC3) in the lamina propria. Anti-inflammatory effects of AGT-5 were also validated on human tonsil cells, where in vitro exposure to AGT-5 increased the proportion of immunosuppressive dendritic cells and ILC3. These results suggest that AGT-5, by stimulating AHR, may promote a general immunosuppressive environment in the pancreas and small intestine lamina propria at the early phase of disease, and thereby inhibit the severity of T1D in mice.
Collapse
Affiliation(s)
- Natalija Jonić
- Department of Immunology, Institute for Biological Research “Siniša Stanković” - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ivan Koprivica
- Department of Immunology, Institute for Biological Research “Siniša Stanković” - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Stavroula G. Kyrkou
- Section of Organic Chemistry & Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Vasileios-Panagiotis Bistas
- Section of Organic Chemistry & Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Christos Chatzigiannis
- Section of Organic Chemistry & Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Nataša Radulović
- Department of Immunology, Institute for Biological Research “Siniša Stanković” - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ivan Pilipović
- Department of Immunology, Institute for Biological Research “Siniša Stanković” - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Andjelina Jovanović
- Department of Otorhinolaryngology with Maxillofacial Surgery, Clinical Hospital Center “Zemun”, Belgrade, Serbia
| | - Milan B. Jovanović
- Department of Otorhinolaryngology with Maxillofacial Surgery, Clinical Hospital Center “Zemun”, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Mirjana Dimitrijević
- Department of Immunology, Institute for Biological Research “Siniša Stanković” - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Andreas G. Tzakos
- Section of Organic Chemistry & Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
- Institute of Materials Science and Computing, University Research Center of Ioannina (URCI), Ioannina, Greece
| | - Ivana Stojanović
- Department of Immunology, Institute for Biological Research “Siniša Stanković” - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
5
|
Kobyliak N, Khomenko M, Falalyeyeva T, Fedchenko A, Savchuk O, Tseyslyer Y, Ostapchenko L. Probiotics for pancreatic β-cell function: from possible mechanism of action to assessment of effectiveness. Crit Rev Microbiol 2024; 50:663-683. [PMID: 37705353 DOI: 10.1080/1040841x.2023.2257776] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 07/27/2023] [Accepted: 09/06/2023] [Indexed: 09/15/2023]
Abstract
Type 2 diabetes (T2D) is a metabolic disease characterized by chronic hyperglycemia because of insulin resistance (IR) and\or pancreatic β-cell dysfunction. Last century research showed that gut microbiota has a direct effect on metabolism and metabolic diseases. New studies into the human microbiome and its connection with the host is making it possible to develop new therapies for a wide variety of diseases. Inflammation is a well-known precursor to metabolic syndrome, which increases the risk of hypertension, visceral obesity, and dyslipidemia, which can lead to T2D through the damage of pancreatic β-cell and reduce insulin secretion. Current understanding for beneficial effects of probiotics in T2D strictly rely on both animal and clinical data, which mostly focused on their impact on IR, anthropometric parameters, glycemic control and markers of chronic systemic inflammation. From the other hand, there is a lack of evidence-based probiotic efficacy on pancreatic β-cell function in terms of T2D and related metabolic disorders. Therefore, current review will focus on the efficacy of probiotics for the protection of β-cells damage and it`s mechanism in patients with T2D.
Collapse
Affiliation(s)
- Nazarii Kobyliak
- Endocrinology Department, Bogomolets National Medical University, Kyiv, Ukraine
- Medical Laboratory CSD, Kyiv, Ukraine
| | - Maria Khomenko
- Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Tetyana Falalyeyeva
- Medical Laboratory CSD, Kyiv, Ukraine
- Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | | | | | | | | |
Collapse
|
6
|
Mihailović M, Soković Bajić S, Arambašić Jovanović J, Brdarić E, Dinić S, Grdović N, Uskoković A, Rajić J, Đorđević M, Tolinački M, Golić N, Živković M, Vidaković M. Beneficial Effects of Probiotic Lactobacillus paraplantarum BGCG11 on Pancreatic and Duodenum Function in Diabetic Rats. Int J Mol Sci 2024; 25:7697. [PMID: 39062940 PMCID: PMC11277547 DOI: 10.3390/ijms25147697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Diabetes mellitus, as a chronic metabolic disorder, significantly impacts the pancreas and among other organs, affects duodenal function. Emerging evidence suggests that probiotics can exert beneficial effects on gut health and metabolism. In our previous research, we evaluated the probiotic Lactobacillus paraplantarum BGCG11 primarily for its protective properties against diabetic rats' damaged liver and kidneys. In this work, we further examined the effects of probiotic strain BGCG11 on the function of the duodenum and pancreas in diabetic rats. We explored the potential mechanisms underlying the probiotic's effects, focusing on general indicators of diabetes, the architecture and morphology of pancreatic islets, duodenal integrity (measuring the transfer of fluid and serum zonulin level), and the modulation of gut microbiota composition. Our findings reveal the protective and regulatory roles of L. paraplantarum BGCG11 in mitigating diabetes-induced pancreatic and duodenal dysfunction regardless of its application time (pre- or post-treatment), highlighting its therapeutic potential in managing diabetes-related gastrointestinal complications.
Collapse
Affiliation(s)
- Mirjana Mihailović
- Institute for Biological Research “Siniša Stanković”, University of Belgrade, Bulevar Despota Stefana 142, 10060 Belgrade, Serbia; (M.M.); (J.A.J.); (S.D.); (N.G.); (A.U.); (J.R.); (M.Đ.)
| | - Svetlana Soković Bajić
- Institute of Molecular Genetics and Genetics Engineering, University of Belgrade, Vojvode Stepe 444a, P.O. Box 23, 11010 Belgrade, Serbia (E.B.); (M.T.); (N.G.)
| | - Jelena Arambašić Jovanović
- Institute for Biological Research “Siniša Stanković”, University of Belgrade, Bulevar Despota Stefana 142, 10060 Belgrade, Serbia; (M.M.); (J.A.J.); (S.D.); (N.G.); (A.U.); (J.R.); (M.Đ.)
| | - Emilija Brdarić
- Institute of Molecular Genetics and Genetics Engineering, University of Belgrade, Vojvode Stepe 444a, P.O. Box 23, 11010 Belgrade, Serbia (E.B.); (M.T.); (N.G.)
| | - Svetlana Dinić
- Institute for Biological Research “Siniša Stanković”, University of Belgrade, Bulevar Despota Stefana 142, 10060 Belgrade, Serbia; (M.M.); (J.A.J.); (S.D.); (N.G.); (A.U.); (J.R.); (M.Đ.)
| | - Nevena Grdović
- Institute for Biological Research “Siniša Stanković”, University of Belgrade, Bulevar Despota Stefana 142, 10060 Belgrade, Serbia; (M.M.); (J.A.J.); (S.D.); (N.G.); (A.U.); (J.R.); (M.Đ.)
| | - Aleksandra Uskoković
- Institute for Biological Research “Siniša Stanković”, University of Belgrade, Bulevar Despota Stefana 142, 10060 Belgrade, Serbia; (M.M.); (J.A.J.); (S.D.); (N.G.); (A.U.); (J.R.); (M.Đ.)
| | - Jovana Rajić
- Institute for Biological Research “Siniša Stanković”, University of Belgrade, Bulevar Despota Stefana 142, 10060 Belgrade, Serbia; (M.M.); (J.A.J.); (S.D.); (N.G.); (A.U.); (J.R.); (M.Đ.)
| | - Marija Đorđević
- Institute for Biological Research “Siniša Stanković”, University of Belgrade, Bulevar Despota Stefana 142, 10060 Belgrade, Serbia; (M.M.); (J.A.J.); (S.D.); (N.G.); (A.U.); (J.R.); (M.Đ.)
| | - Maja Tolinački
- Institute of Molecular Genetics and Genetics Engineering, University of Belgrade, Vojvode Stepe 444a, P.O. Box 23, 11010 Belgrade, Serbia (E.B.); (M.T.); (N.G.)
| | - Nataša Golić
- Institute of Molecular Genetics and Genetics Engineering, University of Belgrade, Vojvode Stepe 444a, P.O. Box 23, 11010 Belgrade, Serbia (E.B.); (M.T.); (N.G.)
| | - Milica Živković
- Institute of Molecular Genetics and Genetics Engineering, University of Belgrade, Vojvode Stepe 444a, P.O. Box 23, 11010 Belgrade, Serbia (E.B.); (M.T.); (N.G.)
| | - Melita Vidaković
- Institute for Biological Research “Siniša Stanković”, University of Belgrade, Bulevar Despota Stefana 142, 10060 Belgrade, Serbia; (M.M.); (J.A.J.); (S.D.); (N.G.); (A.U.); (J.R.); (M.Đ.)
| |
Collapse
|
7
|
Sikiric P, Sever M, Krezic I, Vranes H, Kalogjera L, Smoday IM, Vukovic V, Oroz K, Coric L, Skoro M, Kavelj I, Zubcic S, Sikiric S, Beketic Oreskovic L, Oreskovic I, Blagaic V, Brcic K, Strbe S, Staresinic M, Boban Blagaic A, Skrtic A, Seiwerth S. New studies with stable gastric pentadecapeptide protecting gastrointestinal tract. significance of counteraction of vascular and multiorgan failure of occlusion/occlusion-like syndrome in cytoprotection/organoprotection. Inflammopharmacology 2024:10.1007/s10787-024-01499-8. [PMID: 38980576 DOI: 10.1007/s10787-024-01499-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 05/28/2024] [Indexed: 07/10/2024]
Abstract
Since the early 1990s, when Robert's and Szabo's cytoprotection concept had already been more than one decade old, but still not implemented in therapy, we suggest the stable gastric pentadecapeptide BPC 157 as the most relevant mediator of the cytoprotection concept. Consequently, it can translate stomach and gastrointestinal mucosal maintenance, epithelium, and endothelium cell protection to the therapy of other tissue healing (organoprotection), easily applicable, as native and stable in human gastric juice for more than 24 h. These overwhelm current clinical evidence (i.e., ulcerative colitis, phase II, no side effects, and no lethal dose (LD1) in toxicology studies), as BPC 157 therapy effectively combined various tissue healing and lesions counteraction. BPC 157 cytoprotection relevance and vascular recovery, activation of collateral pathways, membrane stabilizer, eye therapy, wound healing capability, brain-gut and gut-brain functioning, tumor cachexia counteraction, muscle, tendon, ligament, and bone disturbances counteraction, and the heart disturbances, myocardial infarction, heart failure, pulmonary hypertension, arrhythmias, and thrombosis counteraction appeared in the recent reviews. Here, as concept resolution, we review the counteraction of advanced Virchow triad circumstances by activation of the collateral rescuing pathways, depending on injury, activated azygos vein direct blood flow delivery, to counteract occlusion/occlusion-like syndromes starting with the context of alcohol-stomach lesions. Counteraction of major vessel failure (congested inferior caval vein and superior mesenteric vein, collapsed azygos vein, collapsed abdominal aorta) includes counteraction of the brain (intracerebral and intraventricular hemorrhage), heart (congestion, severe arrhythmias), lung (hemorrhage), and congestion and lesions in the liver, kidney, and gastrointestinal tract, intracranial (superior sagittal sinus), portal and caval hypertension, aortal hypotension, and thrombosis, peripherally and centrally.
Collapse
Affiliation(s)
- Predrag Sikiric
- Department of Pharmacology, School of Medicine, University of Zagreb, Zagreb, Croatia.
| | - Marko Sever
- Department of Pharmacology, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Surgery, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ivan Krezic
- Department of Pharmacology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Hrvoje Vranes
- Department of Pharmacology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Luka Kalogjera
- Department of Pharmacology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ivan Maria Smoday
- Department of Pharmacology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Vlasta Vukovic
- Department of Pharmacology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Katarina Oroz
- Department of Pharmacology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Luka Coric
- Department of Pharmacology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Marija Skoro
- Department of Pharmacology, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Diagnostic and Interventional Radiology, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia
| | - Ivana Kavelj
- Department of Pharmacology, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Diagnostic and Interventional Radiology, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia
| | - Slavica Zubcic
- Department of Pharmacology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Suncana Sikiric
- Department of Pathology, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia
| | | | - Ivana Oreskovic
- Department of Pharmacology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Vladimir Blagaic
- Department of Pharmacology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Klara Brcic
- Department of Pharmacology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Sanja Strbe
- Department of Pharmacology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Mario Staresinic
- Department of Pharmacology, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Surgery, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Alenka Boban Blagaic
- Department of Pharmacology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Anita Skrtic
- Department of Pathology, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia
| | - Sven Seiwerth
- Department of Pathology, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia
| |
Collapse
|
8
|
Ronen D, Rokach Y, Abedat S, Qadan A, Daana S, Amir O, Asleh R. Human Gut Microbiota in Cardiovascular Disease. Compr Physiol 2024; 14:5449-5490. [PMID: 39109979 DOI: 10.1002/cphy.c230012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
The gut ecosystem, termed microbiota, is composed of bacteria, archaea, viruses, protozoa, and fungi and is estimated to outnumber human cells. Microbiota can affect the host by multiple mechanisms, including the synthesis of metabolites and toxins, modulating inflammation and interaction with other organisms. Advances in understanding commensal organisms' effect on human conditions have also elucidated the importance of this community for cardiovascular disease (CVD). This effect is driven by both direct CV effects and conditions known to increase CV risk, such as obesity, diabetes mellitus (DM), hypertension, and renal and liver diseases. Cardioactive metabolites, such as trimethylamine N -oxide (TMAO), short-chain fatty acids (SCFA), lipopolysaccharides, bile acids, and uremic toxins, can affect atherosclerosis, platelet activation, and inflammation, resulting in increased CV incidence. Interestingly, this interaction is bidirectional with microbiota affected by multiple host conditions including diet, bile acid secretion, and multiple diseases affecting the gut barrier. This interdependence makes manipulating microbiota an attractive option to reduce CV risk. Indeed, evolving data suggest that the benefits observed from low red meat and Mediterranean diet consumption can be explained, at least partially, by the changes that these diets may have on the gut microbiota. In this article, we depict the current epidemiological and mechanistic understanding of the role of microbiota and CVD. Finally, we discuss the potential therapeutic approaches aimed at manipulating gut microbiota to improve CV outcomes. © 2024 American Physiological Society. Compr Physiol 14:5449-5490, 2024.
Collapse
Affiliation(s)
- Daniel Ronen
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yair Rokach
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Suzan Abedat
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Abed Qadan
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Samar Daana
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Offer Amir
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rabea Asleh
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
9
|
Yang X, Huang J, Peng J, Wang P, Wong FS, Wang R, Wang D, Wen L. Gut microbiota from B-cell-specific TLR9-deficient NOD mice promote IL-10 + Breg cells and protect against T1D. Front Immunol 2024; 15:1413177. [PMID: 38903498 PMCID: PMC11187306 DOI: 10.3389/fimmu.2024.1413177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 05/22/2024] [Indexed: 06/22/2024] Open
Abstract
Introduction Type 1 diabetes (T1D) is an autoimmune disease characterized by the destruction of insulin-producing β cells. Toll-like receptor 9 (TLR9) plays a role in autoimmune diseases, and B cell-specific TLR9 deficiency delays T1D development. Gut microbiota are implicated in T1D, although the relationship is complex. However, the impact of B cell-specific deficiency of TLR9 on intestinal microbiota and the impact of altered intestinal microbiota on the development of T1D are unclear. Objectives This study investigated how gut microbiota and the intestinal barrier contribute to T1D development in B cell-specific TLR9-deficient NOD mice. Additionally, this study explored the role of microbiota in immune regulation and T1D onset. Methods The study assessed gut permeability, gene expression related to gut barrier integrity, and gut microbiota composition. Antibiotics depleted gut microbiota, and fecal samples were transferred to germ-free mice. The study also examined IL-10 production, Breg cell differentiation, and their impact on T1D development. Results B cell-specific TLR9-deficient NOD mice exhibited increased gut permeability and downregulated gut barrier-related gene expression. Antibiotics restored gut permeability, suggesting microbiota influence. Altered microbiota were enriched in Lachnospiraceae, known for mucin degradation. Transferring this microbiota to germ-free mice increased gut permeability and promoted IL-10-expressing Breg cells. Rag-/- mice transplanted with fecal samples from Tlr9 fl/fl Cd19-Cre+ mice showed delayed diabetes onset, indicating microbiota's impact. Conclusion B cell-specific TLR9 deficiency alters gut microbiota, increasing gut permeability and promoting IL-10-expressing Breg cells, which delay T1D. This study uncovers a link between TLR9, gut microbiota, and immune regulation in T1D, with implications for microbiota-targeted T1D therapies.
Collapse
Affiliation(s)
- Xin Yang
- Department of Food Science and Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
- Section of Endocrinology, Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| | - Juan Huang
- Section of Endocrinology, Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| | - Jian Peng
- Section of Endocrinology, Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| | - Pai Wang
- Section of Endocrinology, Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
- Department of Gastrocolorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - F. Susan Wong
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Ruirui Wang
- Shanghai Innovation Center of Traditional Chinese Medicine (TCM) Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dapeng Wang
- Department of Food Science and Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Li Wen
- Section of Endocrinology, Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
10
|
Bharadiya V, Rong Y, Zhang Z, Lin R, Guerrerio AL, Tse CM, Donowitz M, Singh V. Type 1 diabetes human enteroid studies reveal major changes in the intestinal epithelial compartment. Sci Rep 2024; 14:11911. [PMID: 38789719 PMCID: PMC11126659 DOI: 10.1038/s41598-024-62282-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Lack of understanding of the pathophysiology of gastrointestinal (GI) complications in type 1 diabetes (T1D), including altered intestinal transcriptomes and protein expression represents a major gap in the management of these patients. Human enteroids have emerged as a physiologically relevant model of the intestinal epithelium but establishing enteroids from individuals with long-standing T1D has proven difficult. We successfully established duodenal enteroids using endoscopic biopsies from pediatric T1D patients and compared them with aged-matched enteroids from healthy subjects (HS) using bulk RNA sequencing (RNA-seq), and functional analyses of ion transport processes. RNA-seq analysis showed significant differences in genes and pathways associated with cell differentiation and proliferation, cell fate commitment, and brush border membrane. Further validation of these results showed higher expression of enteroendocrine cells, and the proliferating cell marker Ki-67, significantly lower expression of NHE3, lower epithelial barrier integrity, and higher fluid secretion in response to cAMP and elevated calcium in T1D enteroids. Enteroids established from pediatric T1D duodenum identify characteristics of an abnormal intestinal epithelium and are distinct from HS. Our data supports the use of pediatric enteroids as an ex-vivo model to advance studies of GI complications and drug discovery in T1D patients.
Collapse
Affiliation(s)
- Vishwesh Bharadiya
- Divisions of Gastroenterology and Hepatology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Yan Rong
- Divisions of Gastroenterology and Hepatology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Zixin Zhang
- Divisions of Gastroenterology and Hepatology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Ruxian Lin
- Divisions of Gastroenterology and Hepatology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | | | - C Ming Tse
- Divisions of Gastroenterology and Hepatology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Mark Donowitz
- Divisions of Gastroenterology and Hepatology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Varsha Singh
- Divisions of Gastroenterology and Hepatology, Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
11
|
Schirone L, Overi D, Carpino G, Carnevale R, De Falco E, Nocella C, D’Amico A, Bartimoccia S, Cammisotto V, Castellani V, Frati G, Sciarretta S, Gaudio E, Pignatelli P, Alvaro D, Violi F. Oleuropein, a Component of Extra Virgin Olive Oil, Improves Liver Steatosis and Lobular Inflammation by Lipopolysaccharides-TLR4 Axis Downregulation. Int J Mol Sci 2024; 25:5580. [PMID: 38891768 PMCID: PMC11171925 DOI: 10.3390/ijms25115580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Gut-dysbiosis-induced lipopolysaccharides (LPS) translocation into systemic circulation has been suggested to be implicated in nonalcoholic fatty liver disease (NAFLD) pathogenesis. This study aimed to assess if oleuropein (OLE), a component of extra virgin olive oil, lowers high-fat-diet (HFD)-induced endotoxemia and, eventually, liver steatosis. An immunohistochemistry analysis of the intestine and liver was performed in (i) control mice (CTR; n = 15), (ii) high-fat-diet fed (HFD) mice (HFD; n = 16), and (iii) HFD mice treated with 6 µg/day of OLE for 30 days (HFD + OLE, n = 13). The HFD mice developed significant liver steatosis compared to the controls, an effect that was significantly reduced in the HFD + OLE-treated mice. The amount of hepatocyte LPS localization and the number of TLR4+ macrophages were higher in the HFD mice in the than controls and were lowered in the HFD + OLE-treated mice. The number of CD42b+ platelets was increased in the liver sinusoids of the HFD mice compared to the controls and decreased in the HFD + OLE-treated mice. Compared to the controls, the HFD-treated mice showed a high percentage of intestine PAS+ goblet cells, an increased length of intestinal crypts, LPS localization and TLR4+ expression, and occludin downregulation, an effect counteracted in the HFD + OLE-treated mice. The HFD-fed animals displayed increased systemic levels of LPS and zonulin, but they were reduced in the HFD + OLE-treated animals. It can be seen that OLE administration improves liver steatosis and inflammation in association with decreased LPS translocation into the systemic circulation, hepatocyte localization of LPS and TLR4 downregulation in HFD-induced mouse model of NAFLD.
Collapse
Affiliation(s)
- Leonardo Schirone
- IRCCS Neuromed, 86077 Pozzilli, Italy; (L.S.); (R.C.); (G.F.); (S.S.)
| | - Diletta Overi
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, Sapienza University of Rome, 00185 Rome, Italy; (D.O.); (G.C.); (E.G.)
| | - Guido Carpino
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, Sapienza University of Rome, 00185 Rome, Italy; (D.O.); (G.C.); (E.G.)
| | - Roberto Carnevale
- IRCCS Neuromed, 86077 Pozzilli, Italy; (L.S.); (R.C.); (G.F.); (S.S.)
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (E.D.F.); (A.D.); (S.B.)
| | - Elena De Falco
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (E.D.F.); (A.D.); (S.B.)
| | - Cristina Nocella
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy; (C.N.); (V.C.); (P.P.)
| | - Alessandra D’Amico
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (E.D.F.); (A.D.); (S.B.)
| | - Simona Bartimoccia
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (E.D.F.); (A.D.); (S.B.)
| | - Vittoria Cammisotto
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy; (C.N.); (V.C.); (P.P.)
| | - Valentina Castellani
- Department of General Surgery and Surgical Speciality Paride Stefanini, Sapienza University of Rome, 00185 Rome, Italy;
| | - Giacomo Frati
- IRCCS Neuromed, 86077 Pozzilli, Italy; (L.S.); (R.C.); (G.F.); (S.S.)
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (E.D.F.); (A.D.); (S.B.)
| | - Sebastiano Sciarretta
- IRCCS Neuromed, 86077 Pozzilli, Italy; (L.S.); (R.C.); (G.F.); (S.S.)
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (E.D.F.); (A.D.); (S.B.)
| | - Eugenio Gaudio
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, Sapienza University of Rome, 00185 Rome, Italy; (D.O.); (G.C.); (E.G.)
| | - Pasquale Pignatelli
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy; (C.N.); (V.C.); (P.P.)
| | - Domenico Alvaro
- Department of Precision and Translational Medicine, Sapienza University of Rome, 00185 Rome, Italy;
| | - Francesco Violi
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy; (C.N.); (V.C.); (P.P.)
| |
Collapse
|
12
|
Neiva LP, Lopez LC, Pasiani RO, Serra MJR, Rullo VEV. Use of probiotics and similar in pediatric patients with Type 1 Diabetes Mellitus: a systematic review. REVISTA PAULISTA DE PEDIATRIA : ORGAO OFICIAL DA SOCIEDADE DE PEDIATRIA DE SAO PAULO 2024; 42:e2023097. [PMID: 38359319 PMCID: PMC10868513 DOI: 10.1590/1984-0462/2024/42/2023097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 09/21/2023] [Indexed: 02/17/2024]
Abstract
OBJECTIVE To perform a systematic review of randomized controlled trials, evaluating the effect of probiotics, prebiotics or symbiotics supplementation on glycemic and inflammatory control in children with Type 1 Diabetes Mellitus (T1DM). DATA SOURCE The Medical Literature Analysis and Retrieval System Online (MEDLINE/PubMed), Clinical Trials, Literatura Latino-Americana e do Caribe em Ciências da Saúde (LILACS) and Scientific Electronic Library Online (SciELO) databases were searched. Randomized clinical trials of pediatric patients with DM1 using probiotics, prebiotics or symbiotics were included, regardless of year or language of publication. Studies that did not evaluate glycated hemoglobin (HbA1c) were excluded. Metabolic results (HbA1c, total insulin dose and C-peptide) and inflammatory control [interleukin-10 (IL-10), tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ)] during probiotic supplementation or similar, related to modification of the intestinal microbiota, were analyzed. PROSPERO ID: CRD42022384485. DATA SYNTHESIS Five studies were selected for a systematic review. Regarding metabolic markers, only one of the articles that analyzed HbA1c showed a significant decrease (p=0.03) in the intervention group. One study identified a reduction in the total dose of insulin and increased C-peptide levels. Regarding the evaluation of inflammatory parameters (IL-10, TNF-α, INF-γ), there were no statistical relevant modifications. CONCLUSIONS Current data from the literature were not conclusive in identifying an improvement in glycemic control and did not observe changes in inflammatory parameters with the use of probiotics, prebiotics or symbiotics in pediatric patients with T1DM.
Collapse
|
13
|
Atkinson MA, Mirmira RG. The pathogenic "symphony" in type 1 diabetes: A disorder of the immune system, β cells, and exocrine pancreas. Cell Metab 2023; 35:1500-1518. [PMID: 37478842 PMCID: PMC10529265 DOI: 10.1016/j.cmet.2023.06.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 07/23/2023]
Abstract
Type 1 diabetes (T1D) is widely considered to result from the autoimmune destruction of insulin-producing β cells. This concept has been a central tenet for decades of attempts seeking to decipher the disorder's pathogenesis and prevent/reverse the disease. Recently, this and many other disease-related notions have come under increasing question, particularly given knowledge gained from analyses of human T1D pancreas. Perhaps most crucial are findings suggesting that a collective of cellular constituents-immune, endocrine, and exocrine in origin-mechanistically coalesce to facilitate T1D. This review considers these emerging concepts, from basic science to clinical research, and identifies several key remaining knowledge voids.
Collapse
Affiliation(s)
- Mark A Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA.
| | - Raghavendra G Mirmira
- Departments of Medicine and Pediatrics, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
14
|
Shah N, Kulkarni A, Mongad D, Jaani K, Kajale N, Tamahane V, Bhor S, Ladkat D, Khadilkar V, Gondhalekar K, Shouche Y, Khadilkar AV. Pilot Study on Gut Microbiota Profile in Indian Children with Type 1 Diabetes. Indian J Endocrinol Metab 2023; 27:404-409. [PMID: 38107732 PMCID: PMC10723603 DOI: 10.4103/ijem.ijem_22_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 11/03/2022] [Accepted: 11/20/2022] [Indexed: 12/19/2023] Open
Abstract
Background Non-genetic factors like microbial dysbiosis may be contributing to the increasing incidence/progression of type 1 diabetes mellitus (T1DM). Objectives To analyse the gut microbiota profile in Indian children with T1DM and its effect on glycaemic control. Methodology Faecal samples of 29 children with T1DM were collected and faecal microbial DNA was extracted and subjected to 16S rRNA (ribosomal RNA) sequencing and further analysis. Results The dominant phyla in children with T1DM were Firmicutes and Bacteroidetes. Butyrate-producing bacteria Blautia and Ruminococcus showed a significant negative correlation with the glycosylated haemoglobin (HbA1C) levels (p < 0.05). Coprococcus and Propionibacterium were important negative predictors of glycaemic control (p < 0.05). Conclusion Our study suggests that Indian children with T1DM have a distinct gut microbiome taxonomic composition and that short-chain fatty acid-producing bacteria like Ruminococcus and Blautia (butyrate-producing) may play an important role in the glycaemic control of subjects with T1DM.
Collapse
Affiliation(s)
- Nikhil Shah
- Department of Growth and Pediatric Endocrinology, Hirabai Cowasji Jehangir Medical Research Institute, Jehangir Hospital, Pune, Maharashtra, India
- Department of Pediatrics, Cloudnine Hospital, Malad, Mumbai, Maharashtra, India
| | - Abhijit Kulkarni
- National Centre for Cell Science (NCCS), Pune, Maharashtra, India
| | - Dattatray Mongad
- National Centre for Cell Science (NCCS), Pune, Maharashtra, India
| | - Kunal Jaani
- National Centre for Cell Science (NCCS), Pune, Maharashtra, India
| | - Neha Kajale
- Department of Growth and Pediatric Endocrinology, Hirabai Cowasji Jehangir Medical Research Institute, Jehangir Hospital, Pune, Maharashtra, India
| | - Vaishali Tamahane
- Department of Growth and Pediatric Endocrinology, Hirabai Cowasji Jehangir Medical Research Institute, Jehangir Hospital, Pune, Maharashtra, India
| | - Shital Bhor
- Department of Growth and Pediatric Endocrinology, Hirabai Cowasji Jehangir Medical Research Institute, Jehangir Hospital, Pune, Maharashtra, India
| | - Dipali Ladkat
- Department of Growth and Pediatric Endocrinology, Hirabai Cowasji Jehangir Medical Research Institute, Jehangir Hospital, Pune, Maharashtra, India
| | - Vaman Khadilkar
- Department of Growth and Pediatric Endocrinology, Hirabai Cowasji Jehangir Medical Research Institute, Jehangir Hospital, Pune, Maharashtra, India
- Interdisciplinary School of Health Sciences, Savitribai Phule Pune University, Pune, Maharashtra, India
| | - Ketan Gondhalekar
- Department of Growth and Pediatric Endocrinology, Hirabai Cowasji Jehangir Medical Research Institute, Jehangir Hospital, Pune, Maharashtra, India
| | - Yogesh Shouche
- National Centre for Cell Science (NCCS), Pune, Maharashtra, India
| | - Anuradha V. Khadilkar
- Department of Growth and Pediatric Endocrinology, Hirabai Cowasji Jehangir Medical Research Institute, Jehangir Hospital, Pune, Maharashtra, India
- Interdisciplinary School of Health Sciences, Savitribai Phule Pune University, Pune, Maharashtra, India
| |
Collapse
|
15
|
Koivusaari K, Niinistö S, Nevalainen J, Honkanen J, Ruohtula T, Koreasalo M, Ahonen S, Åkerlund M, Tapanainen H, Siljander H, Miettinen ME, Alatossava T, Ilonen J, Vaarala O, Knip M, Virtanen SM. Infant Feeding, Gut Permeability, and Gut Inflammation Markers. J Pediatr Gastroenterol Nutr 2023; 76:822-829. [PMID: 36913717 DOI: 10.1097/mpg.0000000000003756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/15/2023]
Abstract
OBJECTIVES Increased gut permeability and gut inflammation have been linked to the development of type 1 diabetes. Little is known on whether and how intake of different foods is linked to these mechanisms in infancy. We investigated whether the amount of breast milk and intake of other foods are associated with gut inflammation marker concentrations and permeability. METHODS Seventy-three infants were followed from birth to 12 months of age. Their diet was assessed with structured questionnaires and 3-day weighed food records at the age of 3, 6, 9, and 12 months. Gut permeability was assessed with the lactulose/mannitol test and fecal calprotectin and human β-defensin-2 (HBD-2) concentrations were analyzed from stool samples at the age of 3, 6, 9, and 12 months. The associations between foods and gut inflammation marker concentrations and permeability were analyzed using generalized estimating equations. RESULTS Gut permeability and gut inflammation marker concentrations decreased during the first year of life. Intake of hydrolyzed infant formula ( P = 0.003) and intake of fruits and juices ( P = 0.001) were associated with lower intestinal permeability. Intake of fruits and juices ( P < 0.001), vegetables ( P < 0.001), and oats ( P = 0.003) were associated with lower concentrations of HBD-2. Higher intake of breast milk was associated with higher fecal calprotectin concentrations ( P < 0.001), while intake of fruits and juices ( P < 0.001), vegetables ( P < 0.001), and potatoes ( P = 0.007) were associated with lower calprotectin concentrations. CONCLUSIONS Higher intake of breast milk may contribute to higher calprotectin concentration, whereas several complementary foods may decrease gut permeability and concentrations of calprotectin and HBD-2 in infant gut.
Collapse
Affiliation(s)
- Katariina Koivusaari
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- the Department of Food and Nutrition, University of Helsinki, Helsinki, Finland
| | - Sari Niinistö
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Jaakko Nevalainen
- the Faculty of Social Sciences, Unit of Health Sciences, Tampere University, Tampere, Finland
| | - Jarno Honkanen
- the Research Program for Translational Immunology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Terhi Ruohtula
- the Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mirva Koreasalo
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Suvi Ahonen
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Mari Åkerlund
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Heli Tapanainen
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Heli Siljander
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Maija E Miettinen
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Tapani Alatossava
- the Department of Food and Nutrition, University of Helsinki, Helsinki, Finland
| | - Jorma Ilonen
- the Immunogenetics Laboratory, University of Turku, Turku, Finland
| | - Outi Vaarala
- the Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikael Knip
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Suvi M Virtanen
- From the Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| |
Collapse
|
16
|
Saksida T, Paunović V, Koprivica I, Mićanović D, Jevtić B, Jonić N, Stojanović I, Pejnović N. Development of Type 1 Diabetes in Mice Is Associated with a Decrease in IL-2-Producing ILC3 and FoxP3 + Treg in the Small Intestine. Molecules 2023; 28:molecules28083366. [PMID: 37110604 PMCID: PMC10141349 DOI: 10.3390/molecules28083366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/20/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023] Open
Abstract
Recent data indicate the link between the number and function of T regulatory cells (Treg) in the gut immune tissue and initiation and development of autoimmunity associated with type 1 diabetes (T1D). Since type 3 innate lymphoid cells (ILC3) in the small intestine are essential for maintaining FoxP3+ Treg and there are no data about the possible role of ILC3 in T1D pathogenesis, the aim of this study was to explore ILC3-Treg link during the development of T1D. Mature diabetic NOD mice had lower frequencies of IL-2-producing ILC3 and Treg in small intestine lamina propria (SILP) compared to prediabetic NOD mice. Similarly, in multiple low doses of streptozotocin (MLDS)-induced T1D in C57BL/6 mice, hyperglycemic mice exhibited lower numbers of ILC3, IL-2+ ILC3 and Treg in SILP compared to healthy controls. To boost T1D severity, mice were treated with broad-spectrum antibiotics (ABX) for 14 days prior to T1D induction by MLDS. The higher incidence of T1D in ABX-treated mice was associated with significantly lower frequencies of IL-2+ ILC3 and FoxP3+ Treg in SILP compared with mice without ABX treatment. The obtained findings show that the lower proportions of IL-2-expressing ILC3 and FoxP3+ Treg in SILP coincided with diabetes progression and severity.
Collapse
Affiliation(s)
- Tamara Saksida
- Department of Immunology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Verica Paunović
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Pasterova 2, 11000 Belgrade, Serbia
| | - Ivan Koprivica
- Department of Immunology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Dragica Mićanović
- Department of Immunology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Bojan Jevtić
- Department of Immunology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Natalija Jonić
- Department of Immunology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Ivana Stojanović
- Department of Immunology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Nada Pejnović
- Department of Immunology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| |
Collapse
|
17
|
Liang L, Saunders C, Sanossian N. Food, gut barrier dysfunction, and related diseases: A new target for future individualized disease prevention and management. Food Sci Nutr 2023; 11:1671-1704. [PMID: 37051344 PMCID: PMC10084985 DOI: 10.1002/fsn3.3229] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 12/20/2022] [Accepted: 12/28/2022] [Indexed: 03/09/2023] Open
Abstract
Dysfunction of gut barrier is known as "leaky gut" or increased intestinal permeability. Numerous recent scientific evidences showed the association between gut dysfunction and multiple gastrointestinal tract (GI) and non-GI diseases. Research also demonstrated that food plays a crucial role to cause or remedy gut dysfunction related to diseases. We reviewed recent articles from electronic databases, mainly PubMed. The data were based on animal models, cell models, and human research in vivo and in vitro models. In this comprehensive review, our aim focused on the relationship between dietary factors, intestinal permeability dysfunction, and related diseases. This review synthesizes currently available literature and is discussed in three parts: (a) the mechanism of gut barrier and function, (b) food and dietary supplements that may promote gut health, and food or medication that may alter gut function, and (c) a table that organizes the synthesized information by general mechanisms for diseases related to leaky gut/intestinal permeability and associated dietary influences. With future research, dietary intervention could be a new target for individualized disease prevention and management.
Collapse
Affiliation(s)
- Linda Liang
- University of Southern CaliforniaLos AngelesCaliforniaUSA
| | | | - Nerses Sanossian
- Department of NeurologyMedical School of Southern CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
18
|
Igudesman D, Crandell J, Corbin KD, Zaharieva DP, Addala A, Thomas JM, Bulik CM, Pence BW, Pratley RE, Kosorok MR, Maahs DM, Carroll IM, Mayer-Davis EJ. Associations of disordered eating with the intestinal microbiota and short-chain fatty acids among young adults with type 1 diabetes. Nutr Metab Cardiovasc Dis 2023; 33:388-398. [PMID: 36586772 PMCID: PMC9925402 DOI: 10.1016/j.numecd.2022.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 11/05/2022] [Accepted: 11/10/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND AND AIMS Disordered eating (DE) in type 1 diabetes (T1D) includes insulin restriction for weight loss with serious complications. Gut microbiota-derived short chain fatty acids (SCFA) may benefit host metabolism but are reduced in T1D. We evaluated the hypothesis that DE and insulin restriction were associated with reduced SCFA-producing gut microbes, SCFA, and intestinal microbial diversity in adults with T1D. METHODS AND RESULTS We collected stool samples at four timepoints in a hypothesis-generating gut microbiome pilot study ancillary to a weight management pilot in young adults with T1D. 16S ribosomal RNA gene sequencing measured the normalized abundance of SCFA-producing intestinal microbes. Gas-chromatography mass-spectrometry measured SCFA (total, acetate, butyrate, and propionate). The Diabetes Eating Problem Survey-Revised (DEPS-R) assessed DE and insulin restriction. Covariate-adjusted and Bonferroni-corrected generalized estimating equations modeled the associations. COVID-19 interrupted data collection, so models were repeated restricted to pre-COVID-19 data. Data were available for 45 participants at 109 visits, which included 42 participants at 65 visits pre-COVID-19. Participants reported restricting insulin "At least sometimes" at 53.3% of visits. Pre-COVID-19, each 5-point DEPS-R increase was associated with a -0.34 (95% CI -0.56, -0.13, p = 0.07) lower normalized abundance of genus Anaerostipes; and the normalized abundance of Lachnospira genus was -0.94 (95% CI -1.5, -0.42), p = 0.02 lower when insulin restriction was reported "At least sometimes" compared to "Rarely or Never". CONCLUSION DE and insulin restriction were associated with a reduced abundance of SCFA-producing gut microbes pre-COVID-19. Additional studies are needed to confirm these associations to inform microbiota-based therapies in T1D.
Collapse
Affiliation(s)
- Daria Igudesman
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, 27599, USA; AdventHealth Translational Research Institute, Orlando, 32804, USA.
| | - Jamie Crandell
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, 27599, USA
| | - Karen D Corbin
- AdventHealth Translational Research Institute, Orlando, 32804, USA
| | - Dessi P Zaharieva
- Department of Pediatrics, Division of Endocrinology, Stanford University, Stanford, 94304, USA
| | - Ananta Addala
- Department of Pediatrics, Division of Endocrinology, Stanford University, Stanford, 94304, USA
| | - Joan M Thomas
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, 27599, USA
| | - Cynthia M Bulik
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, 27599, USA; Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, USA; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Brian W Pence
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, 27599, USA
| | | | - Michael R Kosorok
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, 27599, USA
| | - David M Maahs
- Department of Pediatrics, Division of Endocrinology, Stanford University, Stanford, 94304, USA
| | - Ian M Carroll
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, 27599, USA
| | - Elizabeth J Mayer-Davis
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, 27599, USA; Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, 27599, USA
| |
Collapse
|
19
|
Niu C, Hu XL, Yuan ZW, Xiao Y, Ji P, Wei YM, Hua YL. Pulsatilla decoction improves DSS-induced colitis via modulation of fecal-bacteria-related short-chain fatty acids and intestinal barrier integrity. JOURNAL OF ETHNOPHARMACOLOGY 2023; 300:115741. [PMID: 36162543 DOI: 10.1016/j.jep.2022.115741] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 08/30/2022] [Accepted: 09/15/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pulsatilla decoction (PD), is an herbal formula commonly used for the treatment of ulcerative colitis (UC) in clinical practice, but the mechanism of PD alters the colitis remains elusive. AIM OF THE STUDY To evaluate the intervention effect of PD on Dextran Sodium Sulfate (DSS)-induced UC based on gut microbiota and intestinal short-chain fatty acid (SCFAs) metabolism, and to investigate the mechanism of action of PD in treating UC. MATERIALS AND METHODS A 3% (wt/vol) DSS-induced ulcerative colitis model in C57BL/6 male mice was used to evaluate the effect of oral PD in treating UC. The changes in gut microbiota in mice were analyzed by 16SrDNA gene sequencing, and the content of SCFAs in the intestinal contents of mice was determined by gas chromatography-mass spectrometry (GC-MS). Enzyme-linked immunosorbent assay (ELISA) was applied to analyze the expression of inflammatory cytokines in serum and colonic tissues, and western blotting (WB) was applied to analyze the expression of tight junction proteins in colonic tissues. RESULTS PD can alleviate the symptoms of UC mice, Pulsatilla Decoction high dose treatment group (PDHT) shows the best effect. Compared with the DSS group, the PDHT had significantly lower body mass, disease activity index (DAI) score, colonic macroscopic damage index (CMDI) score, and pathological damage score, at the phylum level, the relative abundance of Bacteroidetes increased while that of Firmicutes and Proteobacteria decreased, at the Genus level, the abundance of Bacteroides and Lachnospiraceae.NK4A136.group increased while that of Clostridium. sensu.stricto。, Escherichia. shigella and Turicibacter decreased. Compared with the DSS group, acetate, propionate, and total SCFAs in the PDHT with significantly higher levels. The concentrations of interleukin-1β (L-1β), tumor necrosis factor-alpha (TNF-α), and interleukin-17 (IL-17) decreased whereby the concentration of interleukin-10 (IL-10) increased in the PDHT group. The expression levels of Occludin, zonula occludens-1 (ZO-1), Claudin1, Claudin5, G protein-coupled receptor43 (GPR43) protein, and the relative expression of ZO-1 and Occludin mRNA were significantly increased PDHT group. CONCLUSIONS PD has a good therapeutic effect on UC mice. The pharmacological mechanism is probably maintaining the homeostasis and diversity of gut microbiota, increasing the content of SCFAs, and repairing the colonic mucosal barrier.
Collapse
Affiliation(s)
- Chun Niu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, PR China.
| | - Xiao-Li Hu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, PR China.
| | - Zi-Wen Yuan
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, PR China.
| | - Ying Xiao
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, PR China.
| | - Peng Ji
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, PR China.
| | - Yan-Ming Wei
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, PR China.
| | - Yong-Li Hua
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, PR China.
| |
Collapse
|
20
|
Abstract
Systemic inflammation has been suggested to have a pivotal role in atherothrombosis, but the factors that trigger systemic inflammation have not been fully elucidated. Lipopolysaccharide (LPS) is a component of the membrane of Gram-negative bacteria present in the gut that can translocate into the systemic circulation, causing non-septic, low-grade endotoxaemia. Gut dysbiosis is a major determinant of low-grade endotoxaemia via dysfunction of the intestinal barrier scaffold, which is a prerequisite for LPS translocation into the systemic circulation. Experimental studies have demonstrated that LPS is present in atherosclerotic arteries but not in normal arteries. In atherosclerotic plaques, LPS promotes a pro-inflammatory status that can lead to plaque instability and thrombus formation. Low-grade endotoxaemia affects several cell types, including leukocytes, platelets and endothelial cells, leading to inflammation and clot formation. Low-grade endotoxaemia has been described in patients at risk of or with overt cardiovascular disease, in whom low-grade endotoxaemia was associated with atherosclerotic burden and its clinical sequelae. In this Review, we describe the mechanisms favouring the development of low-grade endotoxaemia, focusing on gut dysbiosis and changes in gut permeability; the plausible biological mechanisms linking low-grade endotoxaemia and atherothrombosis; the clinical studies suggesting that low-grade endotoxaemia is a risk factor for cardiovascular events; and the potential therapeutic tools to improve gut permeability and eventually eliminate low-grade endotoxaemia.
Collapse
|
21
|
Rampanelli E, Nieuwdorp M. Gut microbiome in type 1 diabetes: the immunological perspective. Expert Rev Clin Immunol 2023; 19:93-109. [PMID: 36401835 DOI: 10.1080/1744666x.2023.2150612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
INTRODUCTION Type 1 diabetes (T1D) is a prevalent, and yet uncurable, autoimmune disease targeting insulin-producing pancreatic β-cells. Despite a known genetic component in T1D onset, genetics alone cannot explain the alarming worldwide rise in T1D incidence, which is attributed to a growing impact of environmental factors, including perturbations of the gut microbiome. AREAS COVERED Intestinal commensal bacteria plays a crucial role in host physiology in health and disease by regulating endocrine and immune functions. An aberrant gut microbiome structure and metabolic function have been documented prior and during T1D onset. In this review, we summarize and discuss the current studies depicting the taxonomic profile and role of the gut microbial communities in murine models of T1D, diabetic patients and human interventional trials. EXPERT OPINION Compelling evidence have shown that the intestinal microbiota is instrumental in driving differentiation and functions of immune cells. Therefore, any alterations in the intestinal microbiome composition or microbial metabolite production, particularly early in life, may impact disease susceptibility and amplify inflammatory responses and hence accelerate the course of T1D pathogenesis.
Collapse
Affiliation(s)
- Elena Rampanelli
- Department of Experimental Vascular Medicine, Amsterdam University Medical Center, Location AMC, Amsterdam, The Netherlands.,Amsterdam Institute for Infection and Immunity (AII), Amsterdam, The Netherlands.,Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM) Institute, Amsterdam, The Netherlands.,Amsterdam Cardiovascular Sciences (ACS) Institute, Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM) Institute, Amsterdam, The Netherlands.,Amsterdam Cardiovascular Sciences (ACS) Institute, Amsterdam, The Netherlands.,Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, Amsterdam, The Netherlands
| |
Collapse
|
22
|
Alka Ahuja, Saraswathy Mp, Nandakumar S, Prakash F A, Kn G, Um D. Role of the Gut Microbiome in Diabetes and Cardiovascular Diseases Including Restoration and Targeting Approaches- A Review. DRUG METABOLISM AND BIOANALYSIS LETTERS 2022; 15:133-149. [PMID: 36508273 DOI: 10.2174/2949681015666220615120300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/15/2022] [Accepted: 03/18/2022] [Indexed: 12/15/2022]
Abstract
Metabolic diseases, including cardiovascular diseases (CVD) and diabetes, have become the leading cause of morbidity and mortality worldwide. Gut microbiota appears to play a vital role in human disease and health, according to recent scientific reports. The gut microbiota plays an important role in sustaining host physiology and homeostasis by creating a cross-talk between the host and microbiome via metabolites obtained from the host's diet. Drug developers and clinicians rely heavily on therapies that target the microbiota in the management of metabolic diseases, and the gut microbiota is considered the biggest immune organ in the human body. They are highly associated with intestinal immunity and systemic metabolic disorders like CVD and diabetes and are reflected as potential therapeutic targets for the management of metabolic diseases. This review discusses the mechanism and interrelation between the gut microbiome and metabolic disorders. It also highlights the role of the gut microbiome and microbially derived metabolites in the pathophysiological effects related to CVD and diabetes. It also spotlights the reasons that lead to alterations of microbiota composition and the prominence of gut microbiota restoration and targeting approaches as effective treatment strategies in diabetes and CVD. Future research should focus onunderstanding the functional level of some specific microbial pathways that help maintain physiological homeostasis, multi-omics, and develop novel therapeutic strategies that intervene with the gut microbiome for the prevention of CVD and diabetes that contribute to a patient's well-being.
Collapse
Affiliation(s)
- Alka Ahuja
- College of Pharmacy, National University of Science and Technology, PC130, Muscat, Sultanate of Oman
| | - Saraswathy Mp
- Department of Microbiology, ESIC Medical College and PGIMSR, Chennai-600078, India
| | - Nandakumar S
- Department of Biotechnology, Pondicherry University, Kalapet, Puducherry-605014, India
| | - Arul Prakash F
- Centre of Molecular Medicine and Diagnostics (COMMAND), Saveetha Dental College and Hospital, Saveetha Institute of Medical & Technical Sciences, Chennai- 600077, India
| | - Gurpreet Kn
- College of Pharmacy, National University of Science and Technology, PC130, Muscat, Sultanate of Oman
| | - Dhanalekshmi Um
- College of Pharmacy, National University of Science and Technology, PC130, Muscat, Sultanate of Oman
| |
Collapse
|
23
|
Urlacher SS, Kim EY, Luan T, Young LJ, Adjetey B. Minimally invasive biomarkers in human and non-human primate evolutionary biology: Tools for understanding variation and adaptation. Am J Hum Biol 2022; 34:e23811. [PMID: 36205445 PMCID: PMC9787651 DOI: 10.1002/ajhb.23811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/21/2022] [Accepted: 09/10/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The use of minimally invasive biomarkers (MIBs - physiological biomarkers obtained from minimally invasive sample types) has expanded rapidly in science and medicine over the past several decades. The MIB approach is a methodological strength in the field of human and non-human primate evolutionary biology (HEB). Among humans and our closest relatives, MIBs provide unique opportunities to document phenotypic variation and to operationalize evolutionary hypotheses. AIMS This paper overviews the use of MIBs in HEB. Our objectives are to (1) highlight key research topics which successfully implement MIBs, (2) identify promising yet under-investigated areas of MIB application, and (3) discuss current challenges in MIB research, with suggestions for advancing the field. DISCUSSION AND CONCLUSIONS A range of MIBs are used to investigate focal topics in HEB, including energetics and life history variation/evolution, developmental plasticity, and social status and dominance relationships. Nonetheless, we identify gaps in existing MIB research on traits such as physical growth and gut function that are central to the field. Several challenges remain for HEB research using MIBs, including the need for additional biomarkers and methods of assessment, robust validations, and approaches that are standardized across labs and research groups. Importantly, researchers must provide better support for adaptation and fitness effects in hypothesis testing (e.g., by obtaining complementary measures of energy expenditure, demonstrating redundancy of function, and performing lifetime/longitudinal analyses). We point to continued progress in the use of MIBs in HEB to better understand the past, present, and future of humans and our closest primate relatives.
Collapse
Affiliation(s)
- Samuel S. Urlacher
- Department of AnthropologyBaylor UniversityWacoTexasUSA
- Human Evolutionary Biology and Health LabBaylor UniversityWacoTexasUSA
- Child and Brain Development ProgramCIFARTorontoOntarioCanada
| | - Elizabeth Y. Kim
- Human Evolutionary Biology and Health LabBaylor UniversityWacoTexasUSA
- Department of BiologyBaylor UniversityWacoTexasUSA
| | - Tiffany Luan
- Human Evolutionary Biology and Health LabBaylor UniversityWacoTexasUSA
| | - Lauren J. Young
- Human Evolutionary Biology and Health LabBaylor UniversityWacoTexasUSA
| | - Brian Adjetey
- Human Evolutionary Biology and Health LabBaylor UniversityWacoTexasUSA
| |
Collapse
|
24
|
Syromyatnikov M, Nesterova E, Gladkikh M, Smirnova Y, Gryaznova M, Popov V. Characteristics of the Gut Bacterial Composition in People of Different Nationalities and Religions. Microorganisms 2022; 10:microorganisms10091866. [PMID: 36144468 PMCID: PMC9501501 DOI: 10.3390/microorganisms10091866] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 11/25/2022] Open
Abstract
High-throughput sequencing has made it possible to extensively study the human gut microbiota. The links between the human gut microbiome and ethnicity, religion, and race remain rather poorly understood. In this review, data on the relationship between gut microbiota composition and the nationality of people and their religion were generalized. The unique gut microbiome of a healthy European (including Slavic nationality) is characterized by the dominance of the phyla Firmicutes, Bacteroidota, Actinobacteria, Proteobacteria, Fusobacteria, and Verrucomicrobia. Among the African population, the typical members of the microbiota are Bacteroides and Prevotella. The gut microbiome of Asians is very diverse and rich in members of the genera Prevotella, Bacteroides Lactobacillus, Faecalibacterium, Ruminococcus, Subdoligranulum, Coprococcus, Collinsella, Megasphaera, Bifidobacterium, and Phascolarctobacterium. Among Buddhists and Muslims, the Prevotella enterotype is characteristic of the gut microbiome, while other representatives of religions, including Christians, have the Bacteroides enterotype. Most likely, the gut microbiota of people of different nationalities and religions are influenced by food preferences. The review also considers the influences of pathologies such as obesity, Crohn’s disease, cancer, diabetes, etc., on the bacterial composition of the guts of people of different nationalities.
Collapse
Affiliation(s)
- Mikhail Syromyatnikov
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia
- Correspondence:
| | - Ekaterina Nesterova
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia
| | - Maria Gladkikh
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia
| | - Yuliya Smirnova
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia
| | - Mariya Gryaznova
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia
| | - Vasily Popov
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia
| |
Collapse
|
25
|
He L, Chen R, Zhang B, Zhang S, Khan BA, Zhu D, Wu Z, Xiao C, Chen B, Chen F, Hou K. Fecal microbiota transplantation treatment of autoimmune-mediated type 1 diabetes mellitus. Front Immunol 2022; 13:930872. [PMID: 36032108 PMCID: PMC9414079 DOI: 10.3389/fimmu.2022.930872] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/11/2022] [Indexed: 02/05/2023] Open
Abstract
UNLABELLED Type 1 diabetes mellitus (T1DM) is an autoimmune-mediated disease characterized by a reduced or absolute lack of insulin secretion and often associated with a range of vascular and neurological complications for which there is a lack of effective treatment other than lifestyle interventions and pharmacological treatments such as insulin injections. Studies have shown that the gut microbiota is involved in mediating the onset and development of many fecal and extrafecal diseases, including autoimmune T1DM. In recent years, many cases of gut microbiota transplantation for diseases of the bowel and beyond have been reported worldwide, and this approach has been shown to be safe and effective. Here, we conducted an experimental treatment study in two adolescent patients diagnosed with autoimmune T1DM for one year. Patients received one to three rounds of normal fecal microbiota transplants (FMT) and were followed for up to 30 weeks. Clinical outcomes were measured, including biochemical indices, medication regimen, and dosage adjustment. Fecal microbiota metagenomic sequencing after transplantation provides a reference for more reasonable and effective microbiota transplantation protocols to treat autoimmune T1DM. Our results suggest that FMT is an effective treatment for autoimmune T1DM. CLINICAL TRIAL REGISTRATION http://www.chictr.org.cn, identifier ChiCTR2100045789.
Collapse
Affiliation(s)
- Lina He
- Department of Endocrine and Metabolic Diseases, Longhu People’s Hospital, Shantou, China
- Key Laboratory for Research on Active Ingredients in Natural Medicine of Jiangxi Province, Yichun University, Yichun, China
| | - Rongping Chen
- School of Laboratory Medical and Biotechnology, Southern Medical University, Guangzhou, China
| | - Bangzhou Zhang
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- School of Basic Medical Science, Central South University, Changsha, China
| | - Shuo Zhang
- Department of Endocrine and Metabolic Diseases, Longhu People’s Hospital, Shantou, China
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Barkat Ali Khan
- Drug Delivery and Cosmetics Lab, Good Clinical Practice (GCPS), Faculty of Pharmacy, Gomal University, Dera Ismail Khan, Pakistan
| | - Dan Zhu
- Department of Endocrine and Metabolic Diseases, Longhu People’s Hospital, Shantou, China
| | - Zezhen Wu
- Department of Endocrine and Metabolic Diseases, Longhu People’s Hospital, Shantou, China
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Chuanxing Xiao
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- School of Basic Medical Science, Central South University, Changsha, China
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Baolong Chen
- Center for Research and Development, Xiamen Treatgut Biotechnology Co. Ltd., Xiamen, China
| | - Fengwu Chen
- Department of Endocrine and Metabolic Diseases, Longhu People’s Hospital, Shantou, China
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Kaijian Hou
- Department of Endocrine and Metabolic Diseases, Longhu People’s Hospital, Shantou, China
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
26
|
Garabatos N, Santamaria P. Gut Microbial Antigenic Mimicry in Autoimmunity. Front Immunol 2022; 13:873607. [PMID: 35572569 PMCID: PMC9094498 DOI: 10.3389/fimmu.2022.873607] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/14/2022] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota plays a major role in the developmental biology and homeostasis of cells belonging to the adaptive and innate arms of the immune system. Alterations in its composition, which are known to be regulated by both genetic and environmental factors, can either promote or suppress the pathogenic processes underlying the development of various autoimmune diseases, including inflammatory bowel disease, multiple sclerosis, systemic lupus erythematosus, type 1 diabetes and rheumatoid arthritis, to just name a few. Cross-recognition of gut microbial antigens by autoreactive T cells as well as gut microbe-driven alterations in the activation and homeostasis of effector and regulatory T cells have been implicated in this process. Here, we summarize our current understanding of the positive and negative associations between alterations in the composition of the gut microbiota and the development of various autoimmune disorders, with a special emphasis on antigenic mimicry.
Collapse
Affiliation(s)
- Nahir Garabatos
- Institut D'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Pere Santamaria
- Institut D'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Julia McFarlane Diabetes Research Centre (JMDRC), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
27
|
Could I-FABP Be an Early Marker of Celiac Disease in Children with Type 1 Diabetes? Retrospective Study from the Tertiary Reference Centre. Nutrients 2022; 14:nu14030414. [PMID: 35276772 PMCID: PMC8840733 DOI: 10.3390/nu14030414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/09/2022] [Accepted: 01/16/2022] [Indexed: 02/05/2023] Open
Abstract
Patients with type 1 diabetes (T1D) are at higher risk of celiac disease (CD). Recently, intestinal fatty acid binding protein (I-FABP) has been shown to be a serological biomarker of impaired intestinal barrier in CD. Thus, the aim of this study was to verify whether I-FABP could be an early marker of CD in pediatric T1D patients. I-FABP was measured in sera of patients with T1D (n = 156), active CD (n = 38), T1D with active CD (T1D-CD, n= 51), and age-matched healthy children (n = 55). Additionally, I-FABP was determined in T1D patients with negative CD serology at least one year before CD diagnosis (T1D-CD-1, n = 22), in CD patients on a gluten-free diet (CD-GFD, n = 36), and T1D-CD patients on GFD (T1D-CD-GFD, n = 39). Sera were tested using immunoenzymatic assay. Significantly increased levels of I-FABP were found in the T1D, active CD, and T1D-CD groups (1153 ± 665, 1104 ± 916, and 1208 ± 878, respectively) in comparison to healthy with controls (485 ± 416, p < 0.05). GFD induced a significant decrease in I-FABP levels in CD and T1D-CD groups (510 ± 492 and 548 ± 439, respectively). Interestingly, in T1D-CD-1 and T1D, I-FABP levels were comparable (833 ± 369 vs. 1153 ± 665), and significantly increased in relation to healthy controls and T1D-CD values on GFD. The results indicate that the epithelial barrier is disrupted in T1D patients independently of CD development; therefore, I-FABP cannot serve as an early marker of CD in T1D patients. Although GFD can improve epithelial recovery, the question remains as to whether GFD could exert beneficial effects on the intestinal barrier in early stages of T1D.
Collapse
|
28
|
Infection with the enteric pathogen C. rodentium promotes islet-specific autoimmunity by activating a lymphatic route from the gut to pancreatic lymph node. Mucosal Immunol 2022; 15:471-479. [PMID: 35140345 PMCID: PMC9038524 DOI: 10.1038/s41385-022-00490-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 02/04/2023]
Abstract
In nonobese diabetic (NOD) mice, C. rodentium promotes priming of islet-specific T-cells in pancreatic lymph nodes (PaLN), which is a critical step in initiation and perpetuation of islet-autoimmunity. To investigate mechanisms by which C. rodentium promotes T-cell priming in PaLN, we used fluorescent imaging of lymphatic vasculature emanating from colon, followed dendritic cell (DC) migration from colon using photoconvertible-reporter mice, and evaluated the translocation of bacteria to lymph nodes with GFP-C. rodentium and in situ hybridization of bacterial DNA. Fluorescent dextran injected in the colon wall accumulated under subcapsular sinus of PaLN indicating the existence of a lymphatic route from colon to PaLN. Infection with C. rodentium induced DC migration from colon to PaLN and bacterial DNA was detected in medullary sinus and inner cortex of PaLN. Following infection with GFP-C. rodentium, fluorescence appeared in macrophages and gut-derived (CD103+) and resident (CD103-/XCR1+) DC, indicating transportation of bacteria from colon to PaLN both by DC and by lymph itself. This induced proinflammatory cytokine transcripts, activation of DC and islet-specific T-cells in PaLN of NOD mice. Our findings demonstrate the existence of a direct, enteric pathogen-activated route for lymph, cells, and bacteria from colon, which promotes activation of islet-specific T-cells in PaLN.
Collapse
|
29
|
Kühn F, Duan R, Ilmer M, Wirth U, Adiliaghdam F, Schiergens TS, Andrassy J, Bazhin AV, Werner J. Targeting the Intestinal Barrier to Prevent Gut-Derived Inflammation and Disease: A Role for Intestinal Alkaline Phosphatase. Visc Med 2021; 37:383-393. [PMID: 34722721 DOI: 10.1159/000515910] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/16/2021] [Indexed: 02/02/2023] Open
Abstract
Background Intestinal alkaline phosphatase (IAP) as a tissue-specific isozyme of alkaline phosphatases is predominantly produced by enterocytes in the proximal small intestine. In recent years, an increasing number of pathologies have been identified to be associated with an IAP deficiency, making it very worthwhile to review the various roles, biological functions, and potential therapeutic aspects of IAP. Summary IAP primarily originates and acts in the intestinal tract but affects other organs through specific biological axes related to its fundamental roles such as promoting gut barrier function, dephosphorylation/detoxification of lipopolysaccharides (LPS), and regulation of gut microbiota. Key Messages Numerous studies reporting on the different roles and the potential therapeutic value of IAP across species have been published during the last decade. While IAP deficiency is linked to varying degrees of physiological dysfunctions across multiple organ systems, the supplementation of IAP has been proven to be beneficial in several translational and clinical studies. The increasing evidence of the salutary functions of IAP underlines the significance of the naturally occurring brush border enzyme.
Collapse
Affiliation(s)
- Florian Kühn
- Department of General, Visceral and Transplant Surgery, University Hospital of LMU Munich, Munich, Germany
| | - Ruifeng Duan
- Department of General, Visceral and Transplant Surgery, University Hospital of LMU Munich, Munich, Germany
| | - Matthias Ilmer
- Department of General, Visceral and Transplant Surgery, University Hospital of LMU Munich, Munich, Germany
| | - Ulrich Wirth
- Department of General, Visceral and Transplant Surgery, University Hospital of LMU Munich, Munich, Germany
| | - Fatemeh Adiliaghdam
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tobias S Schiergens
- Department of General, Visceral and Transplant Surgery, University Hospital of LMU Munich, Munich, Germany
| | - Joachim Andrassy
- Department of General, Visceral and Transplant Surgery, University Hospital of LMU Munich, Munich, Germany
| | - Alexandr V Bazhin
- Department of General, Visceral and Transplant Surgery, University Hospital of LMU Munich, Munich, Germany
| | - Jens Werner
- Department of General, Visceral and Transplant Surgery, University Hospital of LMU Munich, Munich, Germany
| |
Collapse
|
30
|
Siljander H, Jason E, Ruohtula T, Selvenius J, Koivusaari K, Salonen M, Ahonen S, Honkanen J, Ilonen J, Vaarala O, Virtanen SM, Lähdeaho ML, Knip M. Effect of Early Feeding on Intestinal Permeability and Inflammation Markers in Infants with Genetic Susceptibility to Type 1 Diabetes: A Randomized Clinical Trial. J Pediatr 2021; 238:305-311.e3. [PMID: 34293372 DOI: 10.1016/j.jpeds.2021.07.042] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 06/16/2021] [Accepted: 07/15/2021] [Indexed: 12/21/2022]
Abstract
OBJECTIVES To assess whether weaning to an extensively hydrolyzed formula (EHF) decreases gut permeability and/or markers of intestinal inflammation in infants with HLA-conferred diabetes susceptibility, when compared with conventional formula. STUDY DESIGN By analyzing 1468 expecting biological parent pairs for HLA-conferred susceptibility for type 1 diabetes, 465 couples (32 %) potentially eligible for the study were identified. After further parental consent, 332 babies to be born were randomized at 35th gestational week. HLA genotyping was performed at birth in 309 infants. Out of 87 eligible children, 73 infants participated in the intervention study: 33 in the EHF group and 40 in the control group. Clinical visits took place at 3, 6, 9, and 12 months of age. The infants were provided either EHF or conventional formula whenever breastfeeding was not available or additional feeding was required over the first 9 months of life. The main outcome was the lactulose to mannitol ratio (L/M ratio) at 9 months. The secondary outcomes were L/M ratio at 3, 6, and 12 months of age, and fecal calprotectin and human beta-defensin 2 (HBD-2) levels at each visit. RESULTS Compared with controls, the median L/M ratio was lower in the EHF group at 9 months (.006 vs .028; P = .005). Otherwise, the levels of intestinal permeability, fecal calprotectin, and HBD-2 were comparable between the two groups, although slight differences in the age-related dynamics of these markers were observed. CONCLUSIONS It is possible to decrease intestinal permeability in infancy through weaning to an extensively hydrolyzed formula. This may reduce the early exposure to dietary antigens. TRIAL REGISTRATION Clinicaltrials.gov: NCT01735123.
Collapse
Affiliation(s)
- Heli Siljander
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Eeva Jason
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Center for Child Health Research, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Terhi Ruohtula
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Clinicum, University of Helsinki, Helsinki, Finland
| | - Jenni Selvenius
- Center for Child Health Research, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Katariina Koivusaari
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland; Department of Food and Nutrition, University of Helsinki, Helsinki, Finland
| | - Marja Salonen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Suvi Ahonen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland; Tampere University Hospital, Research, Development and Innovation Center, Tampere, Finland; Unit of Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Jarno Honkanen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Clinicum, University of Helsinki, Helsinki, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, University of Turku and Turku University of Hospital, Turku, Finland
| | - Outi Vaarala
- Clinicum, University of Helsinki, Helsinki, Finland
| | - Suvi M Virtanen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland; Tampere University Hospital, Research, Development and Innovation Center, Tampere, Finland; Unit of Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Marja-Leena Lähdeaho
- Center for Child Health Research, University of Tampere and Tampere University Hospital, Tampere, Finland; Department of Pediatrics, Tampere University Hospital, Tampere, Finland
| | - Mikael Knip
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Center for Child Health Research, University of Tampere and Tampere University Hospital, Tampere, Finland; Department of Pediatrics, Tampere University Hospital, Tampere, Finland.
| |
Collapse
|
31
|
Morse ZJ, Horwitz MS. Virus Infection Is an Instigator of Intestinal Dysbiosis Leading to Type 1 Diabetes. Front Immunol 2021; 12:751337. [PMID: 34721424 PMCID: PMC8554326 DOI: 10.3389/fimmu.2021.751337] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 09/28/2021] [Indexed: 12/12/2022] Open
Abstract
In addition to genetic predisposition, environmental determinants contribute to a complex etiology leading to onset of type 1 diabetes (T1D). Multiple studies have established the gut as an important site for immune modulation that can directly impact development of autoreactive cell populations against pancreatic self-antigens. Significant efforts have been made to unravel how changes in the microbiome function as a contributor to autoimmune responses and can serve as a biomarker for diabetes development. Large-scale longitudinal studies reveal that common environmental exposures precede diabetes pathology. Virus infections, particularly those associated with the gut, have been prominently identified as risk factors for T1D development. Evidence suggests recent-onset T1D patients experience pre-existing subclinical enteropathy and dysbiosis leading up to development of diabetes. The start of these dysbiotic events coincide with detection of virus infections. Thus viral infection may be a contributing driver for microbiome dysbiosis and disruption of intestinal homeostasis prior to T1D onset. Ultimately, understanding the cross-talk between viral infection, the microbiome, and the immune system is key for the development of preventative measures against T1D.
Collapse
Affiliation(s)
| | - Marc S. Horwitz
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
32
|
Otuya DO, Gavgiotaki E, Carlson CJ, Shi SQ, Lee AJ, Krall AA, Chung A, Grant CG, Bhat NM, Choy P, Giddings SL, Gardecki JA, Thiagarajah JR, Rowe SM, Tearney GJ. Minimally Invasive Image-Guided Gut Transport Function Measurement Probe. FRONTIERS IN PHYSICS 2021; 9:735645. [PMID: 36382063 PMCID: PMC9648666 DOI: 10.3389/fphy.2021.735645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
INTRODUCTION Diseases such as celiac disease, environmental enteric dysfunction, infectious gastroenteritis, type II diabetes and inflammatory bowel disease are associated with increased gut permeability. Dual sugar absorption tests, such as the lactulose to rhamnose ratio (L:R) test, are the current standard for measuring gut permeability. Although easy to administer in adults, the L:R test has a number of drawbacks. These include an inability to assess for spatial heterogeneity in gut permeability that may distinguish different disease severity or pathology, additional sample collection for immunoassays, and challenges in carrying out the test in certain populations such as infants and small children. Here, we demonstrate a minimally invasive probe for real-time localized gut permeability evaluation through gut potential difference (GPD) measurement. MATERIALS AND METHODS The probe has an outer diameter of 1.2 mm diameter and can be deployed in the gut of unsedated subjects via a transnasal introduction tube (TNIT) that is akin to an intestinal feeding tube. The GPD probe consists of an Ag/AgCl electrode, an optical probe and a perfusion channel all housed within a transparent sheath. Lactated Ringer's (LR) solution is pumped through the perfusion channel to provide ionic contact between the electrodes and the gut lining. The optical probe captures non-scanning (M-mode) OCT images to confirm electrode contact with the gut lining. A separate skin patch probe is placed over an abraded skin area to provide reference for the GPD measurements. Swine studies were conducted to validate the GPD probe. GPD in the duodenum was modulated by perfusing 45 ml of 45 mM glucose. RESULTS GPD values of -13.1 ± 2.8 mV were measured in the duodenum across four swine studies. The change in GPD in the duodenum with the addition of glucose was -10.5 ± 2.4 mV (p < 0.001). M-mode OCT images provided electrode-tissue contact information, which was vital in ascertaining the probe's proximity to the gut mucosa. CONCLUSION We developed and demonstrated a minimally invasive method for investigating gastrointestinal permeability consisting of an image guided GPD probe that can be used in unsedated subjects.
Collapse
Affiliation(s)
- David O. Otuya
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Evangelia Gavgiotaki
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Camella J. Carlson
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, United States
| | - Serena Q. Shi
- University of Pennsylvania, Philadelphia, MA, United States
| | - Ariel J. Lee
- Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Alexander A. Krall
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, United States
| | - Anita Chung
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, United States
| | - Catriona G. Grant
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, United States
| | - Nitasha M. Bhat
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, United States
| | - Peter Choy
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, United States
| | - Sarah L. Giddings
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, United States
| | - Joseph A. Gardecki
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Jay R. Thiagarajah
- Harvard Medical School, Boston, MA, United States
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Boston, MA, United States
| | - Steven M. Rowe
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Gregory Fleming James Cystic Fibrosis Research Center, Birmingham, AL, United States
| | - Guillermo J. Tearney
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Department of Pathology, Massachusetts General Hospital, Boston, MA, United States
- Harvard-MIT Division of Health Sciences and Technology (HST), Boston, MA, United States
| |
Collapse
|
33
|
Cinek O, Kramna L, Odeh R, Alassaf A, Ibekwe MAU, Ahmadov G, Elmahi BME, Mekki H, Lebl J, Abdullah MA. Eukaryotic viruses in the fecal virome at the onset of type 1 diabetes: A study from four geographically distant African and Asian countries. Pediatr Diabetes 2021; 22:558-566. [PMID: 33786936 DOI: 10.1111/pedi.13207] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/21/2020] [Accepted: 02/26/2021] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Studies of the fecal virome in type 1 diabetes (T1D) have been limited to populations of Europe and the United States. We therefore sought to characterize the stool virome in children after onset of T1D and in matched control subjects from four geographically distant African and Asian countries. METHODS Samples of stool were collected from 73 children and adolescents shortly after T1D onset (Azerbaijan 19, Jordan 20, Nigeria 14, Sudan 20) and 105 matched control subjects of similar age and locale. Metagenomic sequencing of the DNA and RNA virome was performed, and virus positivity was defined as more than 0.001% of reads of the sample. Selected viruses were also quantified using real-time PCR. Conditional logistic regression was used to model associations with eukaryotic virus positivity. RESULTS Signals of 387 different viral species were detected; at least one eukaryotic virus was detected in 71% case and 65% control samples. Neither of observed eukaryotic virus species or genera differed in frequency between children with T1D and controls. There was a suggestive association of the total count of different viral genera per sample between cases (1.45 genera) and controls (1.10 genera, OR 1.24, 95%CI 0.98-1.57), and an unplanned subanalysis suggested marginally more frequent endogenous retrovirus signal in cases (in 28.8% vs. in 8.6% controls, OR = 4.55, 95%CI 1.72-12). CONCLUSIONS No clear and consistent association with T1D was observed in the fecal viromes from four distant non-European populations. The finding of borderline associations of human endogenous retroviruses merits further exploration.
Collapse
Affiliation(s)
- Ondrej Cinek
- Department of Pediatrics, Second Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| | - Lenka Kramna
- Department of Pediatrics, Second Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| | - Rasha Odeh
- Department of Pediatrics, School of Medicine, University of Jordan, Amman, Jordan
| | - Abeer Alassaf
- Department of Pediatrics, School of Medicine, University of Jordan, Amman, Jordan
| | - Mary Ann Ugochi Ibekwe
- Department of Pediatrics, Federal Teaching Hospital Abakaliki, Ebonyi State University, Abakaliki, Nigeria
| | | | - Bashir Mukhtar Elwasila Elmahi
- Department of Paediatrics and Child Health, Faculty of Medicine, University of Khartoum, Khartoum, Sudan.,Sudan Childhood Diabetes Center, Khartoum, Sudan
| | - Hanan Mekki
- Department of Paediatrics and Child Health, Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | - Jan Lebl
- Department of Pediatrics, Second Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| | - Mohammed Ahmed Abdullah
- Department of Paediatrics and Child Health, Faculty of Medicine, University of Khartoum, Khartoum, Sudan.,Sudan Childhood Diabetes Center, Khartoum, Sudan
| |
Collapse
|
34
|
Babu SN, Govindarajan S, Vijayalakshmi MA, Noor A. Role of zonulin and GLP-1/DPP-IV in alleviation of diabetes mellitus by peptide/polypeptide fraction of Aloe vera in streptozotocin- induced diabetic wistar rats. JOURNAL OF ETHNOPHARMACOLOGY 2021; 272:113949. [PMID: 33610707 DOI: 10.1016/j.jep.2021.113949] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/09/2021] [Accepted: 02/14/2021] [Indexed: 06/12/2023]
Abstract
ETHNO-PHARMACOLOGICAL RELEVANCE The genus Aloe has a long history of usage in medicine. Aloe barbadensis Miller, commonly known as Aloe vera, is said to possess anti-diabetic, anti-inflammatory, anti-cancer, anti-microbial, immunomodulation, wound healing properties. AIM OF THE STUDY In diabetes mellitus, loss in intestinal permeability is observed with high levels of zonulin and low levels of glucagon-like peptide-1 (GLP-1) leading to hyperglycemia. The aim of the study was to understand the role of peptide/polypeptide fraction (PPF) of Aloe vera in the alleviation of diabetes through maintaining the intestinal permeability by regulating the zonulin and GLP-1 levels. MATERIALS AND METHODS The PPF of Aloe vera was obtained through trichloroacetic acid precipitation. The anti-diabetic potential of the PPF was tested through DPP-IV inhibition, glucose diffusion assay, and by using Rin-m5F cells. The anti-diabetic potential of the PPF was tested at a dose of 0.450 mg/kg bw in vivo using streptozotocin-induced diabetic Wistar rats. The effect of PPF on fasting plasma glucose, insulin, glucagon, Zonulin, GLP-1, DPP-IV, levels were studied in diabetic rats. The histopathological studies of the pancreas, small intestine, and liver were carried out for organ-specific effects. RESULTS PPF has the ability to reduce fasting plasma glucose levels with concomitant increase in insulin levels in streptozotocin-induced diabetic rats. It was also observed that increase in GLP-1 levels with a decrease in DPP-IV and zonulin levels thereby mitigating the loss of intestinal permeability. These findings correlate with the small intestine's histopathological observation where the excessive proliferation of epithelium in the small intestine of diabetic rats was reduced after PPF treatment. CONCLUSION These results suggest that the PPF of Aloe vera alleviates diabetes through islet cell rejuvenation via GLP-1/DPP-IV pathway and thereby suggesting the usage of PPF as an alternate medicine for diabetes mellitus with the possibility to reduce the intestinal permeability and zonulin levels.
Collapse
Affiliation(s)
- Spoorthy N Babu
- Centre for Bioseparation Technology, Vellore Institute of Technology, Vellore, 632014, India
| | - S Govindarajan
- Centre for Bioseparation Technology, Vellore Institute of Technology, Vellore, 632014, India
| | - M A Vijayalakshmi
- Centre for Bioseparation Technology, Vellore Institute of Technology, Vellore, 632014, India
| | - Ayesha Noor
- Centre for Bioseparation Technology, Vellore Institute of Technology, Vellore, 632014, India.
| |
Collapse
|
35
|
FFAR from the Gut Microbiome Crowd: SCFA Receptors in T1D Pathology. Metabolites 2021; 11:metabo11050302. [PMID: 34064625 PMCID: PMC8151283 DOI: 10.3390/metabo11050302] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 12/14/2022] Open
Abstract
The gut microbiome has emerged as a novel determinant of type 1 diabetes (T1D), but the underlying mechanisms are unknown. In this context, major gut microbial metabolites, short-chain fatty acids (SCFAs), are considered to be an important link between the host and gut microbiome. We, along with other laboratories, have explored how SCFAs and their cognate receptors affect various metabolic conditions, including obesity, type 2 diabetes, and metabolic syndrome. Though gut microbiome and SCFA-level changes have been reported in T1D and in mouse models of the disease, the role of SCFA receptors in T1D remains under explored. In this review article, we will highlight the existing and possible roles of these receptors in T1D pathology. We conclude with a discussion of SCFA receptors as therapeutic targets for T1D, exploring an exciting new potential for novel treatments of glucometabolic disorders.
Collapse
|
36
|
Lin S, Mukherjee S, Li J, Hou W, Pan C, Liu J. Mucosal immunity-mediated modulation of the gut microbiome by oral delivery of probiotics into Peyer's patches. SCIENCE ADVANCES 2021; 7:7/20/eabf0677. [PMID: 33980483 PMCID: PMC8115924 DOI: 10.1126/sciadv.abf0677] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 03/23/2021] [Indexed: 05/02/2023]
Abstract
Methods capable of maintaining gut microbiota homeostasis to prevent bacterial translocation and infection under external threats are critical for multiple facets of human health but have been rarely reported. Here, we describe the elicitation of mucosal immunity to modulate the gut microbiota by oral delivery of living probiotics into Peyer's patches. Probiotics are individually camouflaged within a yeast membrane, on which the embedded β-glucan can facilitate the phagocytosis of microfold cells that locate in the intestinal epithelium. The delivery of probiotics into lymphoid follicles after oral ingestion promotes robust mucosal immune responses and notably upgrades the production of secretory immunoglobulin A. The provoked immunity positively regulates the gut microflora, which, in turn, retains gut homeostasis and provides defense against environmental attacks. In two murine models of gut barrier impairment, oral administration with camouflaged probiotics effectively prevents the breakdown of intestinal barrier and evidences limited bacterial translocation and systemic inflammation.
Collapse
Affiliation(s)
- Sisi Lin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Subhajit Mukherjee
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Juanjuan Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Weiliang Hou
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Chao Pan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Jinyao Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China.
- Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
37
|
Rüb AM, Tsakmaklis A, Gräfe SK, Simon MC, Vehreschild MJ, Wuethrich I. Biomarkers of human gut microbiota diversity and dysbiosis. Biomark Med 2021; 15:137-148. [PMID: 33442994 DOI: 10.2217/bmm-2020-0353] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 11/04/2020] [Indexed: 12/24/2022] Open
Abstract
The association of gut microbiota dysbiosis with various human diseases is being substantiated with increasing evidence. Metabolites derived from both, microbiota and the human host play a central role in disease susceptibility and disease progression by extensively modulating host physiology and metabolism. Several of these metabolites have the potential to serve as diagnostic biomarkers for monitoring disease states in conjunction with intestinal microbiota dysbiosis. In this narrative review we evaluate the potential of trimethylamine-N-oxide, short-chain fatty acids, 3-indoxyl sulfate, p-cresyl sulfate, secondary bile acids, hippurate, human β-defensin-2, chromogranin A, secreted immunoglobulins and zonulin to serve as biomarkers for metabolite profiling and diagnostic suitability for dysbiosis and disease.
Collapse
Affiliation(s)
- Alina M Rüb
- Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Anastasia Tsakmaklis
- Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Stefanie K Gräfe
- Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Marie-Christine Simon
- Department of Nutrition & Food Sciences, Nutrition & Microbiota, University of Bonn, Bonn, Germany
| | - Maria Jgt Vehreschild
- Department of Internal Medicine, Infectious Diseases, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Irene Wuethrich
- Department of Biosystems Science & Engineering, ETH Zurich, Basel, Switzerland
| |
Collapse
|
38
|
Verduci E, Mameli C, Amatruda M, Petitti A, Vizzuso S, El Assadi F, Zuccotti G, Alabduljabbar S, Terranegra A. Early Nutrition and Risk of Type 1 Diabetes: The Role of Gut Microbiota. Front Nutr 2021; 7:612377. [PMID: 33425976 PMCID: PMC7785819 DOI: 10.3389/fnut.2020.612377] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022] Open
Abstract
Type 1 diabetes (T1D) appears most frequently in childhood, with an alarming increasing incidence in the last decades. Although the genetic predisposition is a major risk factor, it cannot solely explain the complex etiology of T1D which is still not fully understood. In this paper, we reviewed the most recent findings on the role of early nutrition and the involvement of the gut microbiota in the etiopathogenesis of T1D. The main conclusions that are withdrawn from the current literature regarding alleviating the risk of developing T1D through nutrition are the encouragement of long-term breast-feeding for at least the first 6 months of life and the avoidance of early complementary foods and gluten introduction (before 4 months of age) as well as cow milk introduction before 12 months of life. These detrimental feeding habits create a gut microbiota dysbiotic state that can contribute to the onset of T1D in infancy. Finally, we discussed the possibility to introduce probiotics, prebiotics and post-biotics in the prevention of T1D.
Collapse
Affiliation(s)
- Elvira Verduci
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy.,Department of Health Sciences, University of Milan, Milan, Italy
| | - Chiara Mameli
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy
| | - Matilde Amatruda
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Agnese Petitti
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy
| | - Sara Vizzuso
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Farah El Assadi
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Gianvincenzo Zuccotti
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy
| | | | | |
Collapse
|
39
|
Bayrak NA, Volkan B. Helicobacter pylori Infection in Children with Concurrent Celiac Disease and Type 1 Diabetes Mellitus. Dig Dis 2021; 39:444-450. [PMID: 33429389 DOI: 10.1159/000514276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 01/10/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVES Data regarding Helicobacter pylori (Hp) infection frequency in concurrent celiac disease (CD) and type 1 diabetes mellitus (T1DM) (CD + T1DM) are anecdotal. This study aimed to evaluate the association between Hp and concomitant CD + T1DM in children. METHODS In this 2-center, case-control study, children who underwent esophago-gastro-duodenoscopy (EGD) were studied. CD diagnosis was established by favorable histology and serology. Hp infection was confirmed by both histology and the rapid urease test. Patients were divided into 3 groups as CD only (CDo), CD + T1DM, and non-CD children who underwent endoscopy as controls. RESULTS Among the 1,431 EGDs performed, 783 cases were eligible. Overall, 215 cases had CDo (mean age: 9.12 ± 4.18 years, 58.1% girls), 63 cases had CD + T1DM (mean age: 9.29 ± 4.46 years, 50.8% girls), and 505 cases were controls (mean age: 9.69 ± 4.52 years, 56.6% girls). Hp infection rate was significantly lower in CD + T1DM group (controls: 49.7% vs. CDo: 32.1% vs. CD + T1DM: 20.6%, p < 0.01). After adjustment for age, gender, and socioeconomic status, the Hp infection rate was still significantly low (adjusted odds ratio [aOR]: 1.57, 95% confidence interval (CI): 1.35-1.83, p < 0.01). A difference in Hp infection rate between controls and CDo group (aOR: 1.43, 95% CI: 1.09-2.12, p < 0.05), and between CDo and CD + T1DM (aOR: 0.89, 95% CI: 0.65-1.54, p < 0.05) group was significant. The severity of duodenal lesions and the presence of Hp infection were not correlated in all celiac children (r: 0.113, p > 0.05). CONCLUSION The frequency of Hp infection was significantly lower in CD + T1DM children, compared to the CDo group and the controls.
Collapse
Affiliation(s)
- Nevzat Aykut Bayrak
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Diyarbakir Children's Hospital, Diyarbakir, Turkey, .,Division of Pediatric Gastroenterology, Hepatology and Nutrition, University of Health Sciences, Zeynep Kâmil Women & Children's Training & Research Hospital, Istanbul, Turkey,
| | - Burcu Volkan
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Erzurum State Training & Research Hospital, Erzurum, Turkey
| |
Collapse
|
40
|
Park JM, Lee HJ, Sikiric P, Hahm KB. BPC 157 Rescued NSAID-cytotoxicity Via Stabilizing Intestinal Permeability and Enhancing Cytoprotection. Curr Pharm Des 2021; 26:2971-2981. [PMID: 32445447 DOI: 10.2174/1381612826666200523180301] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 04/19/2020] [Indexed: 02/08/2023]
Abstract
The stable gastric pentadecapeptide BPC 157 protects stomach cells, maintains gastric integrity against various noxious agents such as alcohol, nonsteroidal anti-inflammatory drugs (NSAIDs), and exerts cytoprotection/ adaptive cytoprotection/organoprotection in other epithelia, that is, skin, liver, pancreas, heart, and brain. Especially BPC 157 counteracts gastric endothelial injury that precedes and induces damage to the gastric epithelium and generalizes "gastric endothelial protection" to protection of the endothelium of other vessels including thrombosis, prolonged bleeding, and thrombocytopenia. In this background, we put the importance of BPC 157 as a possible way of securing GI safety against NSAIDs-induced gastroenteropathy since still unmet medical needs to mitigate NSAIDs-induced cytotoxicity are urgent. Furthermore, gastrointestinal irritants such as physical or mental stress, NSAIDs administration, surfactants destroyer such as bile acids, alcohol can lead to leaky gut syndrome through increasing epithelial permeability. In this review article, we described the potential rescuing actions of BPC 157 against leaky gut syndrome after NSAIDs administration for the first time.
Collapse
Affiliation(s)
- Jong M Park
- Department of Pharmacology Daejeon University College of Oriental Medicine, Daejeon, Korea
| | - Ho J Lee
- University of Gachon Lee Gil Ya Cancer and Diabetes Institute, Incheon, Korea
| | - Predrag Sikiric
- Department of Pharmacology, Medical Faculty, University of Zagreb, Zagreb, Croatia
| | - Ki B Hahm
- CHA Cancer Prevention Research Center, CHA Bio Complex, Seongnam, Korea.,Digestive Disease Center, CHA University Bundang Medical Center, Pangyo, Korea
| |
Collapse
|
41
|
Hartzler ML, Castle L, Lewis C, Zakaria L. A functional approach to the chronic disease epidemic. Am J Health Syst Pharm 2020; 77:668-672. [PMID: 32236457 DOI: 10.1093/ajhp/zxaa032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Melody L Hartzler
- Department of Pharmacy Practice, Cedarville University School of Pharmacy, Cedarville, OH, and Western Medicine Family, Physicians, Fairborn, OH
| | - Lauren Castle
- Functional Medicine Pharmacists Alliance, Miamisburg, OH
| | - Christine Lewis
- Potter's House Apothecary, Midwestern University College of Pharmacy-Glendale, Glendale, AZ
| | | |
Collapse
|
42
|
Leaky Gut and Autoimmunity: An Intricate Balance in Individuals Health and the Diseased State. Int J Mol Sci 2020; 21:ijms21249770. [PMID: 33371435 PMCID: PMC7767453 DOI: 10.3390/ijms21249770] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/08/2020] [Accepted: 12/17/2020] [Indexed: 02/07/2023] Open
Abstract
Damage to the tissue and the ruining of functions characterize autoimmune syndromes. This review centers around leaky gut syndromes and how they stimulate autoimmune pathogenesis. Lymphoid tissue commonly associated with the gut, together with the neuroendocrine network, collaborates with the intestinal epithelial wall, with its paracellular tight junctions, to maintain the balance, tolerance, and resistance to foreign/neo-antigens. The physiological regulator of paracellular tight junctions plays a vital role in transferring macromolecules across the intestinal barrier and thereby maintains immune response equilibrium. A new paradigm has explained the intricacies of disease development and proposed that the processes can be prevented if the interaction between the genetic factor and environmental causes is barred by re-instituting the intestinal wall function. The latest clinical evidence and animal models reinforce this current thought and offer the basis for innovative methodologies to thwart and treat autoimmune syndromes.
Collapse
|
43
|
Muehler A, Slizgi JR, Kohlhof H, Groeppel M, Peelen E, Vitt D. Clinical relevance of intestinal barrier dysfunction in common gastrointestinal diseases. World J Gastrointest Pathophysiol 2020; 11:114-130. [PMID: 33362939 PMCID: PMC7739114 DOI: 10.4291/wjgp.v11.i6.114] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/07/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023] Open
Abstract
The intestinal barrier is a complex and well-controlled physiological construct designed to separate luminal contents from the bowel wall. In this review, we focus on the intestinal barrier’s relationship with the host’s immune system interaction and the external environment, specifically the microbiome. The bowel allows the host to obtain nutrients vital to survival while protecting itself from harmful pathogens, luminal antigens, or other pro-inflammatory factors. Control over barrier function and the luminal milieu is maintained at the biochemical, cellular, and immunological level. However, disruption to this highly regulated environment can cause disease. Recent advances to the field have progressed the mechanistic understanding of compromised intestinal barrier function in the context of gastrointestinal pathology. There are numerous examples where bowel barrier dysfunction and the resulting interaction between the microbiome and the immune system has disease-triggering consequences. The purpose of this review is to summarize the clinical relevance of intestinal barrier dysfunction in common gastrointestinal and related diseases. This may help highlight the importance of restoring barrier function as a therapeutic mechanism of action in gastrointestinal pathology.
Collapse
|
44
|
Sharma L, Riva A. Intestinal Barrier Function in Health and Disease-Any role of SARS-CoV-2? Microorganisms 2020; 8:E1744. [PMID: 33172188 PMCID: PMC7694956 DOI: 10.3390/microorganisms8111744] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 11/01/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022] Open
Abstract
Alterations in the structure and function of the intestinal barrier play a role in the pathogenesis of a multitude of diseases. During the recent and ongoing coronavirus disease (COVID-19) pandemic, it has become clear that the gastrointestinal system and the gut barrier may be affected by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus, and disruption of barrier functions or intestinal microbial dysbiosis may have an impact on the progression and severity of this new disease. In this review, we aim to provide an overview of current evidence on the involvement of gut alterations in human disease including COVID-19, with a prospective outlook on supportive therapeutic strategies that may be investigated to rescue intestinal barrier functions and possibly facilitate clinical improvement in these patients.
Collapse
Affiliation(s)
- Lakshya Sharma
- Faculty of Life Sciences and Medicine, King’s College London, London SE1 1UL, UK;
| | - Antonio Riva
- Faculty of Life Sciences and Medicine, King’s College London, London SE1 1UL, UK;
- Foundation for Liver Research, Institute of Hepatology, London SE5 9NT, UK
| |
Collapse
|
45
|
Pancreatic Diseases and Microbiota: A Literature Review and Future Perspectives. J Clin Med 2020; 9:jcm9113535. [PMID: 33139601 PMCID: PMC7692447 DOI: 10.3390/jcm9113535] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/28/2020] [Accepted: 10/30/2020] [Indexed: 12/12/2022] Open
Abstract
Gut microbiota represent an interesting worldwide research area. Several studies confirm that microbiota has a key role in human diseases, both intestinal (such as inflammatory bowel disease, celiac disease, intestinal infectious diseases, irritable bowel syndrome) and extra intestinal disorders (such as autism, multiple sclerosis, rheumatologic diseases). Nowadays, it is possible to manipulate microbiota by administering prebiotics, probiotics or synbiotics, through fecal microbiota transplantation in selected cases. In this scenario, pancreatic disorders might be influenced by gut microbiota and this relationship could be an innovative and inspiring field of research. However, data are still scarce and controversial. Microbiota manipulation could represent an important therapeutic strategy in the pancreatic diseases, in addition to standard therapies. In this review, we analyze current knowledge about correlation between gut microbiota and pancreatic diseases, by discussing on the one hand existing data and on the other hand future possible perspectives.
Collapse
|
46
|
Bender C, Rodriguez-Calvo T, Amirian N, Coppieters KT, von Herrath MG. The healthy exocrine pancreas contains preproinsulin-specific CD8 T cells that attack islets in type 1 diabetes. SCIENCE ADVANCES 2020; 6:6/42/eabc5586. [PMID: 33067232 PMCID: PMC7567597 DOI: 10.1126/sciadv.abc5586] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 09/03/2020] [Indexed: 05/03/2023]
Abstract
Preproinsulin (PPI) is presumably a crucial islet autoantigen found in patients with type 1 diabetes (T1D) but is also recognized by CD8+ T cells from healthy individuals. We quantified PPI-specific CD8+ T cells within different areas of the human pancreas from nondiabetic controls, autoantibody-positive donors, and donors with T1D to investigate their role in diabetes development. This spatial cellular quantitation revealed unusually high frequencies of autoreactive CD8+ T cells supporting the hypothesis that PPI is indeed a key autoantigen. To our surprise, PPI-specific CD8+ T cells were already abundantly present in the nondiabetic pancreas, thus questioning the dogma that T1D is caused by defective thymic deletion or systemic immune dysregulation. During T1D development, these cells accumulated in and around islets, indicating that an islet-specific trigger such as up-regulation of major histocompatibility complex class I might be essential to unmask beta cells to the immune system.
Collapse
Affiliation(s)
- Christine Bender
- Center for Type 1 Diabetes Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Teresa Rodriguez-Calvo
- Center for Type 1 Diabetes Research, La Jolla Institute for Immunology, La Jolla, CA, USA
- The Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Institute of Diabetes Research, Munich-Neuherberg, Germany
| | - Natalie Amirian
- Center for Type 1 Diabetes Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Ken T Coppieters
- Global Research Project Management, Novo Nordisk, Måløv, Denmark
| | - Matthias G von Herrath
- Center for Type 1 Diabetes Research, La Jolla Institute for Immunology, La Jolla, CA, USA.
- The Novo Nordisk Research Center Seattle Inc., Seattle, WA, USA
| |
Collapse
|
47
|
Sajid M, Biswas K, Singh H, Negi S. Auto-reactivity against gut bacterial peptides in patients with late-onset diabetes. Autoimmunity 2020; 53:385-393. [PMID: 32924613 DOI: 10.1080/08916934.2020.1818232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The depletion of gut mucosal barrier enables exposure of gut microbes/gut microbial products to the host mucosal immunity which may increase the risk of metabolic/inflammatory disorders. These immune responses can lead to the development of mild autoimmunity to metabolic peptides coming from gut bacteria and may result in metabolic diseases like late-onset diabetes (LOD). In the present study, we identified host sera cross-reactivity with gut bacterial peptides similar to host proteins. The interaction between diabetic sera and gut peptides was detected by enzyme-linked immunosorbent assay (ELISA) and results were confirmed using surface plasmon resonance (SPR). The ELISA assay showed a higher level of serum cross-reactivity in LOD patients as compared to non-diabetic controls against three peptides (P-5, P-9, and P-13). SPR analysis confirmed binding-affinity against P-5 and P-13. Also, a significant correlation was observed between inflammatory markers and P-5. This study demonstrates that gut health is important not only for intestinal diseases but also for several late-onset diseases, like, diabetes.
Collapse
Affiliation(s)
- Mohammad Sajid
- National Institute of Pathology, Indian Council of Medical Research (ICMR), New Delhi, India
| | - Krishna Biswas
- Department of Endocrinology, Vardhman Mahavir Medical College, Safdarjung Hospital, New Delhi, India
| | - Harpreet Singh
- AIIMS Computational Genomics Centre, ISRM Division, Indian Council of Medical Research (ICMR), New Delhi, India
| | - Sapna Negi
- National Institute of Pathology, Indian Council of Medical Research (ICMR), New Delhi, India
| |
Collapse
|
48
|
Kim TK, Lee JC, Im SH, Lee MS. Amelioration of Autoimmune Diabetes of NOD Mice by Immunomodulating Probiotics. Front Immunol 2020; 11:1832. [PMID: 33013834 PMCID: PMC7496355 DOI: 10.3389/fimmu.2020.01832] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 07/08/2020] [Indexed: 01/01/2023] Open
Abstract
Type 1 autoimmune diabetes is an autoimmune disease characterized by specific destruction of pancreatic β-cells producing insulin. Recent studies have shown that gut microbiota and immunity are closely linked to systemic immunity, affecting the balance between pro-inflammatory and regulatory immune responses. Altered gut microbiota may be causally related to the development of immune-mediated diseases, and probiotics have been suggested to have modulatory effects on inflammatory diseases and immune disorders. We studied whether a probiotic combination that has immunomodulatory effects on several inflammatory diseases can reduce the incidence of diabetes in non-obese diabetic (NOD) mice, a classical animal model of human T1D. When Immune Regulation and Tolerance 5 (IRT5), a probiotic combination comprising Lactobacillus acidophilus, Lactobacillus casei, Lactobacillus reuteri, Bifidobacterium bifidium, and Streptococcus thermophiles, was administered 6 times a week for 36 weeks to NOD mice, beginning at 4 weeks of age, the incidence of diabetes was significantly reduced. Insulitis score was also significantly reduced, and β-cell mass was conversely increased by IRT5 administration. IRT5 administration significantly reduced gut permeability in NOD mice. The proportion of total regulatory T cells was not changed by IRT5 administration; however, the proportion of CCR9+ regulatory T (Treg) cells expressing gut-homing receptor was significantly increased in pancreatic lymph nodes (PLNs) and lamina propria of the small intestine (SI-LP). Type 1 T helper (Th1) skewing was reduced in PLNs by IRT5 administration. IRT5 could be a candidate for an effective probiotic combination, which can be safely administered to inhibit or prevent type 1 diabetes (T1D).
Collapse
Affiliation(s)
- Tae Kang Kim
- Department of Internal Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | - Sin-Hyeog Im
- ImmunoBiome. Inc., Pohang, South Korea.,Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, South Korea
| | - Myung-Shik Lee
- Department of Internal Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
49
|
Ilchmann-Diounou H, Menard S. Psychological Stress, Intestinal Barrier Dysfunctions, and Autoimmune Disorders: An Overview. Front Immunol 2020; 11:1823. [PMID: 32983091 PMCID: PMC7477358 DOI: 10.3389/fimmu.2020.01823] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
Autoimmune disorders (ADs) are multifactorial diseases involving, genetic, epigenetic, and environmental factors characterized by an inappropriate immune response toward self-antigens. In the past decades, there has been a continuous rise in the incidence of ADs, which cannot be explained by genetic factors alone. Influence of psychological stress on the development or the course of autoimmune disorders has been discussed for a long time. Indeed, based on epidemiological studies, stress has been suggested to precede AD occurrence and to exacerbate symptoms. Furthermore, compiling data showed that most of ADs are associated with gastrointestinal symptoms, that is, microbiota dysbiosis, intestinal hyperpermeability, and intestinal inflammation. Interestingly, social stress (acute or chronic, in adult or in neonate) is a well-described intestinal disrupting factor. Taken together, those observations question a potential role of stress-induced defect of the intestinal barrier in the onset and/or the course of ADs. In this review, we aim to present evidences supporting the hypothesis for a role of stress-induced intestinal barrier disruption in the onset and/or the course of ADs. We will mainly focus on autoimmune type 1 diabetes, multiple sclerosis and systemic lupus erythematosus, ADs for which we could find sufficient circumstantial data to support this hypothesis. We excluded gastrointestinal (GI) ADs like coeliac disease to privilege ADs not focused on intestinal disorders to avoid confounding factors. Indeed, GIADs are characterized by antibodies directed against intestinal barrier actors.
Collapse
MESH Headings
- Animals
- Autoimmune Diseases/epidemiology
- Autoimmune Diseases/immunology
- Autoimmune Diseases/metabolism
- Autoimmune Diseases/microbiology
- Autoimmunity
- Diabetes Mellitus, Type 1/epidemiology
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/microbiology
- Dysbiosis
- Gastrointestinal Microbiome
- Host-Pathogen Interactions
- Humans
- Intestinal Mucosa/immunology
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/microbiology
- Lupus Erythematosus, Systemic/epidemiology
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/metabolism
- Lupus Erythematosus, Systemic/microbiology
- Multiple Sclerosis/epidemiology
- Multiple Sclerosis/immunology
- Multiple Sclerosis/metabolism
- Multiple Sclerosis/microbiology
- Permeability
- Risk Factors
- Stress, Psychological/epidemiology
- Stress, Psychological/immunology
- Stress, Psychological/metabolism
- Stress, Psychological/microbiology
Collapse
Affiliation(s)
| | - Sandrine Menard
- Neuro-Gastroenterology and Nutrition Team, Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| |
Collapse
|
50
|
Kappler K, Hennet T. Emergence and significance of carbohydrate-specific antibodies. Genes Immun 2020; 21:224-239. [PMID: 32753697 PMCID: PMC7449879 DOI: 10.1038/s41435-020-0105-9] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/14/2020] [Accepted: 07/22/2020] [Indexed: 12/14/2022]
Abstract
Carbohydrate-specific antibodies are widespread among all classes of immunoglobulins. Despite their broad occurrence, little is known about their formation and biological significance. Carbohydrate-specific antibodies are often classified as natural antibodies under the assumption that they arise without prior exposure to exogenous antigens. On the other hand, various carbohydrate-specific antibodies, including antibodies to ABO blood group antigens, emerge after the contact of immune cells with the intestinal microbiota, which expresses a vast diversity of carbohydrate antigens. Here we explore the development of carbohydrate-specific antibodies in humans, addressing the definition of natural antibodies and the production of carbohydrate-specific antibodies upon antigen stimulation. We focus on the significance of the intestinal microbiota in shaping carbohydrate-specific antibodies not just in the gut, but also in the blood circulation. The structural similarity between bacterial carbohydrate antigens and surface glycoconjugates of protists, fungi and animals leads to the production of carbohydrate-specific antibodies protective against a broad range of pathogens. Mimicry between bacterial and human glycoconjugates, however, can also lead to the generation of carbohydrate-specific antibodies that cross-react with human antigens, thereby contributing to the development of autoimmune disorders.
Collapse
Affiliation(s)
| | - Thierry Hennet
- Institute of Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|