1
|
Huang C, Rosolowsky E, Nour MA, Butalia S, Ho J, Mayengbam S, Wang W, Pyke S, Virtanen H, Reimer RA. Prebiotic supplementation in patients with type 1 diabetes: study protocol for a randomised controlled trial in Canada. BMJ Open 2025; 15:e102486. [PMID: 40449951 DOI: 10.1136/bmjopen-2025-102486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/03/2025] Open
Abstract
INTRODUCTION Type 1 diabetes (T1D) mellitus is caused by autoimmune destruction of insulin-producing beta-cells, requiring exogenous insulin to sustain life. Achieving near normal blood glucose levels with insulin, a primary goal of diabetes management, carries a significant risk of hypoglycaemia. There is compelling evidence that an abnormal gut microbiota or dysbiosis can increase intestinal permeability (IP) and contribute to dysglycaemia seen in T1D. Given that prebiotic fibre can mitigate dysbiosis, reduce IP and improve glycaemic control, we hypothesise that microbial changes induced by prebiotics contribute to gut and endocrine adaptations that reduce glucose fluctuations, including less hypoglycaemia. In a pilot study, we showed that in children who had T1D for at least 1 year, a 3-month course of prebiotic fibre significantly reduced the frequency of hypoglycaemia. The prebiotic group had an increase in Bifidobacterium with a moderate improvement in IP. Importantly, the prebiotic group maintained their serum C peptide level (marker of residual beta cell function) while the placebo group saw a drop. Given that preserving endogenous beta cell function in patients with T1D, particularly in the first year of diagnosis, reduces hypoglycaemia and glycaemic variability, we propose to examine the effect of prebiotic supplementation in patients with T1D. METHODS AND ANALYSIS This is a multicentre, randomised, double-blind, placebo-controlled study. Individuals (n=144) with T1D will be randomised 1:1 for 6 months to prebiotic (oligofructose-enriched inulin) or placebo (isocaloric maltodextrin). Participants will have three in-person study visits at baseline, 3 months and 6 months. The primary outcome, frequency of hypoglycaemia, will be determined from continuous glucose monitor (CGM) reports and patient blood glucose logs. Secondary outcomes will include glycaemic variability, time-in-range, glycated haemoglobin, stimulated C peptide, IP, serum inflammatory markers, quality of life and fear of hypoglycaemia ratings, as well as gut microbiome and metabolomics analysis. At 9 months, participant CGM data will be used to assess frequency of hypoglycaemia and glycaemic variability at 3 months postintervention. ETHICS AND DISSEMINATION The study received ethical approval from the University of Calgary Conjoint Health Research Ethics Board (REB21-0852). The University of Alberta subsite was granted ethical approval under the province of Alberta's research ethics reciprocity agreement as a participating site (REB21-0852; pSite00000066). The University of Saskatchewan subsite was granted ethical approval by the Biomedical Research Ethics Board (#4149). Trial findings will be disseminated through peer-reviewed publications and conference presentations. TRIAL REGISTRATION NUMBER clinicaltrials.gov NCT04963777.
Collapse
Affiliation(s)
- Carol Huang
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Elizabeth Rosolowsky
- Department of Pediatrics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Women & Children's Health Research Institute, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Munier A Nour
- College of Nursing, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Sonia Butalia
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Josephine Ho
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Shyamchand Mayengbam
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Weilan Wang
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - Shannon Pyke
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - Heidi Virtanen
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Raylene A Reimer
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
2
|
Han YZ, Wang YZY, Zhu XY, Du BX, Wang YX, Zhang XQ, Jia JM, Liu WJ, Zheng HJ. The gut microbiota and diabetic nephropathy: an observational study review and bidirectional Mendelian randomization study. Trials 2025; 26:101. [PMID: 40122887 PMCID: PMC11931829 DOI: 10.1186/s13063-025-08755-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 01/28/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Earlier studies have implicated a crucial link between diabetic nephropathy (DN) and the gut microbiota (GM) by considering the gut-kidney axis; however, the specific cause-and-effect connections between these processes remain unclear. METHODS To compare changes in the GM between DN patients and control subjects, a review of observational studies was performed. The examination focused on the phylum, family, genus, and species/genus categories. To delve deeper into the cause-effect relationship, instrumental variables for 211 GM taxa (9 phyla, 16 classes, 20 orders, 35 families, and 131 genera), which were eligible for the mbQTL (microbial quantitative trait locus) mapping analysis, were collected from the Genome Wide Association Study (GWAS). A Mendelian randomization investigation was then conducted to gauge their impact on DN susceptibility using data from the European Bioinformatics Institute (EBI) and the FinnGen consortium. The European Bioinformatics Institute data included 1032 DN patients and 451,248 controls, while the FinnGen consortium data consisted of 3283 DN patients and 210,463 controls. Two-sample Mendelian randomization (TSMR) was utilized to determine the link between the GM and DN. The primary method for analysis was the inverse variance weighted (IVW) approach. Moreover, a reverse Mendelian randomization analysis was carried out, and the findings were validated through sensitivity assessments. RESULTS This review examined 11 observational studies that satisfied the inclusion and exclusion criteria. There was a significant difference in the abundance of 144 GM taxa between DN patients and controls. By employing the MR technique, 13 bacteria were pinpointed as having a causal link to DN (including 3 unknown GM taxa). Even after Bonferroni correction, the protective impact of the phylum Proteobacteria and genus Dialister (Sequeira et al. Nat Microbiol. 5:304-313, 2020; Liu et al. EBioMedicine. 90:104527, 2023) and the harmful impact of the genus Akkermansia, family Verrucomicrobiaceae, order Verrucomicrobia and class Verrucomicrobiae on DN remained significant. No noticeable heterogeneity or horizontal pleiotropy was detected in the instrumental variables (IVs). However, reverse MR investigations have failed to reveal any substantial causal relationship between DN and the GM. CONCLUSION Differences in the GM among DN patients and healthy controls are explored in observational studies. We verified the possible connection between certain genetically modified genera and DN, thereby emphasizing the connection between the "gut-kidney" axis and new insights into the GM's role in DN pathogenesis underlying DN. Investigations into this association are necessary, and novel biomarkers for the development of targeted preventive strategies against DN are needed.
Collapse
Affiliation(s)
- Yi Zhen Han
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Zhi Yuan Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xing Yu Zhu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Bo Xuan Du
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yao Xian Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | | | - Jia Meng Jia
- School of Management, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Jing Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| | - Hui Juan Zheng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
3
|
Jacob N, Kumar R. Supplementation of High-Strength Oral Probiotics Improves Immune Regulation and Preserves Beta Cells among Children with New-Onset Type 1 Diabetes Mellitus: A Randomised, Double-Blind Placebo Control Trial: Authors' Reply. Indian J Pediatr 2025; 92:319. [PMID: 39168943 DOI: 10.1007/s12098-024-05244-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 08/08/2024] [Indexed: 08/23/2024]
Affiliation(s)
- Neenu Jacob
- Endocrinology and Diabetes Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Rakesh Kumar
- Endocrinology and Diabetes Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India.
| |
Collapse
|
4
|
Lokesh MN, Kumar R, Jacob N, Sachdeva N, Rawat A, Yadav J, Dayal D. Supplementation of High-Strength Oral Probiotics Improves Immune Regulation and Preserves Beta Cells among Children with New-Onset Type 1 Diabetes Mellitus: A Randomised, Double-Blind Placebo Control Trial. Indian J Pediatr 2025; 92:277-283. [PMID: 38557820 DOI: 10.1007/s12098-024-05074-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 01/24/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVES To investigate the mechanism of glycemic control in children with type 1 diabetes (T1D) following high-strength probiotics supplementation by assessing immune-regulatory markers. METHODS In this single-centre randomised double-blinded placebo-controlled study, children with new-onset T1D on regular insulin therapy were randomised into probiotic or placebo groups with 30 children each. The probiotics group received oral powder of Vivomixx®, and the placebo group received corn starch for six months. The primary outcome parameters included induced T regulatory cells (i-Tregs) percentage, insulin autoantibodies (IAA), insulinoma associated 2 autoantibodies (IA2), glutamic acid decarboxylase autoantibodies (GAD 65) and plasma interleukin-10 (IL-10) levels. The secondary outcome variables were changes in plasma C-peptide levels and glycemic control parameters. RESULTS Twenty-three children in the placebo group and 27 in the probiotic group completed the study. There was a significant increase in the percentage of iTregs (3.40 in the probiotic vs. 2.46 in the placebo group; p = 0.034). Median glycated hemoglobin (HbA1c) levels significantly decreased from 68 mmol/mol (8.35%) in the placebo group to 60 mmol/mol (7.55%) in the probiotic group (p = 0.017). Median C-peptide levels were significantly higher in probiotics (0.72 ng/ml) vs. placebo group (0.11 ng/ml) (p = 0.036). The plasma IL-10 levels significantly increased in the probiotic group after six months of treatment (p = 0.002). CONCLUSIONS The high-strength probiotics improved the immunoregulatory milieu, thereby preserving the beta-cell function and better glycemic control.
Collapse
Affiliation(s)
- M N Lokesh
- Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Rakesh Kumar
- Endocrinology and Diabetes Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Sector 12, Chandigarh, 160012, India.
| | - Neenu Jacob
- Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Naresh Sachdeva
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Amit Rawat
- Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Jaivinder Yadav
- Endocrinology and Diabetes Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Sector 12, Chandigarh, 160012, India
| | - Devi Dayal
- Endocrinology and Diabetes Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Sector 12, Chandigarh, 160012, India
| |
Collapse
|
5
|
Golshany H, Helmy SA, Morsy NFS, Kamal A, Yu Q, Fan L. The gut microbiome across the lifespan: how diet modulates our microbial ecosystem from infancy to the elderly. Int J Food Sci Nutr 2025; 76:95-121. [PMID: 39701663 DOI: 10.1080/09637486.2024.2437472] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/16/2024] [Accepted: 11/28/2024] [Indexed: 12/21/2024]
Abstract
This comprehensive review examines the impact of dietary patterns on gut microbiome composition and diversity from infancy to old age, linking these changes to age-related health outcomes. It investigates how the gut microbiome develops and changes across life stages, focusing on the influence of dietary factors. The review explores how early-life feeding practices, including breastfeeding and formula feeding, shape the infant gut microbiota and have lasting effects. In elderly individuals, alterations in the gut microbiome are associated with increased susceptibility to infections, chronic inflammation, metabolic disorders and cognitive decline. The critical role of diet in modulating the gut microbiome throughout life is emphasised, particularly the potential benefits of probiotics and fortified foods in promoting healthy ageing. By elucidating the mechanisms connecting food systems to gut health, this review provides insights into interventions that could enhance gut microbiome resilience and improve health outcomes across the lifespan.
Collapse
Affiliation(s)
- Hazem Golshany
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- Food Science Department, Faculty of Agriculture, Cairo University, Giza, Egypt
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | | | | | - Aya Kamal
- Food Science Department, Faculty of Agriculture, Cairo University, Giza, Egypt
| | - Qun Yu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Liuping Fan
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- Collaborative Innovation Center of Food Safety & Quality Control, Jiangnan University, Wuxi, China
| |
Collapse
|
6
|
Blok L, Hanssen N, Nieuwdorp M, Rampanelli E. From Microbes to Metabolites: Advances in Gut Microbiome Research in Type 1 Diabetes. Metabolites 2025; 15:138. [PMID: 39997763 PMCID: PMC11857261 DOI: 10.3390/metabo15020138] [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: 02/01/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/26/2025] Open
Abstract
Background: Type 1 diabetes (T1D) is a severe chronic T-cell mediated autoimmune disease that attacks the insulin-producing beta cells of the pancreas. The multifactorial nature of T1D involves both genetic and environmental components, with recent research focusing on the gut microbiome as a crucial environmental factor in T1D pathogenesis. The gut microbiome and its metabolites play an important role in modulating immunity and autoimmunity. In recent years, studies have revealed significant alterations in the taxonomic and functional composition of the gut microbiome associated with the development of islet autoimmunity and T1D. These changes include reduced production of short-chain fatty acids, altered bile acid and tryptophan metabolism, and increased intestinal permeability with consequent perturbations of host (auto)immune responses. Methods/Results: In this review, we summarize and discuss recent observational, mechanistic and etiological studies investigating the gut microbiome in T1D and elucidating the intricate role of gut microbes in T1D pathogenesis. Moreover, we highlight the recent advances in intervention studies targeting the microbiota for the prevention or treatment of human T1D. Conclusions: A deeper understanding of the evolution of the gut microbiome before and after T1D onset and of the microbial signals conditioning host immunity may provide us with essential insights for exploiting the microbiome as a prognostic and therapeutic tool.
Collapse
Affiliation(s)
- Lente Blok
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, 1105 AZ Amsterdam, The Netherlands; (N.H.); (M.N.)
| | - Nordin Hanssen
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, 1105 AZ Amsterdam, The Netherlands; (N.H.); (M.N.)
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, 1105 AZ Amsterdam, The Netherlands; (N.H.); (M.N.)
| | - Elena Rampanelli
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, 1105 AZ Amsterdam, The Netherlands; (N.H.); (M.N.)
- Amsterdam Institute for Infection and Immunity (AII), Amsterdam, The Netherlands
| |
Collapse
|
7
|
Li J, Xie Z, Yang L, Guo K, Zhou Z. The impact of gut microbiome on immune and metabolic homeostasis in type 1 diabetes: Clinical insights for prevention and treatment strategies. J Autoimmun 2025; 151:103371. [PMID: 39883994 DOI: 10.1016/j.jaut.2025.103371] [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: 03/11/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 02/01/2025]
Abstract
Type 1 diabetes (T1D) is a complex disease triggered by a combination of genetic and environmental factors, where abnormal autoimmune responses lead to progressive damage of the pancreatic β cells and severe glucose metabolism disorder. Recent studies have increasingly highlighted the close link between gut microbiota dysbiosis and the development of T1D. This review delves into existing population studies to explore the intricate interactions between the gut microbiota and the immune and metabolic homeostasis in T1D. It summarizes how changes in the structure and function of the gut microbiota are closely associated with the onset and progression of T1D across its natural course and clinical stages. More importantly, based on evidence accumulated from clinical observations and trials, we pioneer the discussion on gut microbiota-based T1D prevention and treatment strategies, this not only enriches our understanding of the complex pathological mechanisms of T1D but also provides potential directions for developing novel prevention and treatment strategies.
Collapse
Affiliation(s)
- Jiaqi Li
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhiguo Xie
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Lin Yang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China.
| | - Keyu Guo
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China.
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
8
|
Kennedy EC, Ross FC, O'Shea CA, Lavelle A, Ross P, Dempsey E, Stanton C, Hawkes CP. Observational study protocol: the faecal microbiome in the acute stage of new-onset paediatric type 1 diabetes in an Irish cohort. BMJ Open 2025; 15:e089206. [PMID: 39890137 PMCID: PMC11784173 DOI: 10.1136/bmjopen-2024-089206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 12/06/2024] [Indexed: 02/03/2025] Open
Abstract
INTRODUCTION Type 1 diabetes (T1D) is an autoimmune-mediated disorder caused by the destruction of pancreatic beta cells. Although there is an underlying genetic predisposition to developing T1D, the trigger is multifactorial and likely includes environmental factors. The intestinal microbiome has been identified as one such factor. Previous studies have illustrated differences in the microbiota of people with T1D compared with healthy controls. This study aims to describe the evolution of the microbiome and metabolome during the first year of clinical T1D, or stage 3 T1D diagnosis, and investigate whether there are differences in the microbiome and metabolome of children who present with and without diabetic ketoacidosis. The study will also explore possible associations between the microbiome, metabolome, glycaemic control and beta cell reserve. METHODS AND ANALYSIS This prospective cohort study will include children with newly diagnosed T1D and sibling controls (n=100, males and females) and their faecal microbiome will be characterised using shotgun metagenomic sequencing at multiple time points during the first year of diagnosis. We will develop a microbial culture biobank based on culturomic studies of stool samples from the healthy controls that will support future investigation. Metabolomic analysis will aim to identify additional biomarkers which may be involved in disease presentation and progression. Through this initial exploratory study, we aim to identify specific microbial biomarkers which may be used as future interventional targets throughout the various stages of T1D progression. ETHICS AND DISSEMINATION This study has been approved by the Clinical Research Ethics Committee of the Cork Teaching Hospitals. Study results will be available to patients with T1D and their families, carers, support networks and microbiome societies and other researchers. TRIAL REGISTRATION NUMBER The clinicaltrials.gov registration number for this trial is NCT06157736.
Collapse
Affiliation(s)
- Elaine Catherine Kennedy
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
- INFANT Research Centre, University College Cork, Cork, Ireland
| | - Fiona Catherine Ross
- Department of Anatomy & Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | - Aonghus Lavelle
- Department of Anatomy & Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Eugene Dempsey
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
- INFANT Research Centre, University College Cork, Cork, Ireland
- Department of Neonatology, Cork University Maternity Hospital, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Food Research Centre Moorepark, Moorepark, Ireland
| | - Colin Patrick Hawkes
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
- INFANT Research Centre, University College Cork, Cork, Ireland
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
9
|
Sundheim B, Hirani K, Blaschke M, Lemos JRN, Mittal R. Pre-Type 1 Diabetes in Adolescents and Teens: Screening, Nutritional Interventions, Beta-Cell Preservation, and Psychosocial Impacts. J Clin Med 2025; 14:383. [PMID: 39860389 PMCID: PMC11765808 DOI: 10.3390/jcm14020383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/19/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
Type 1 Diabetes (T1D) is a progressive autoimmune disease often identified in childhood or adolescence, with early stages detectable through pre-diabetic markers such as autoantibodies and subclinical beta-cell dysfunction. The identification of the pre-T1D stage is critical for preventing complications, such as diabetic ketoacidosis, and for enabling timely interventions that may alter disease progression. This review examines the multifaceted approach to managing T1D risk in adolescents and teens, emphasizing early detection, nutritional interventions, beta-cell preservation strategies, and psychosocial support. Screening for T1D-associated autoantibodies offers predictive insight into disease risk, particularly when combined with education and family resources that promote lifestyle adjustments. Although nutritional interventions alone are not capable of preventing T1D, certain lifestyle interventions, such as weight management and specific nutritional choices, have shown the potential to preserve insulin sensitivity, reduce inflammation, and mitigate metabolic strain. Pharmacological strategies, including immune-modulating drugs like teplizumab, alongside emerging regenerative and cell-based therapies, offer the potential to delay disease onset by protecting beta-cell function. The social and psychological impacts of a T1D risk diagnosis are also significant, affecting adolescents' quality of life, family dynamics, and mental health. Supportive interventions, including counseling, cognitive-behavioral therapy (CBT), and group support, are recommended for managing the emotional burden of pre-diabetes. Future directions call for integrating universal or targeted screening programs within schools or primary care, advancing research into nutrition and psychosocial support, and promoting policies that enhance access to preventive resources. Advocacy for the insurance coverage of screening, nutritional counseling, and mental health services is also crucial to support families in managing T1D risk. By addressing these areas, healthcare systems can promote early intervention, improve beta-cell preservation, and support the overall well-being of adolescents at risk of T1D.
Collapse
Affiliation(s)
- Brody Sundheim
- Young Leaders Advocacy Group, Diabetes Research Institute Foundation, Hollywood, FL 33021, USA; (B.S.); (K.H.); (M.B.); (J.R.N.L.)
- Ransom Everglades High School, 3575 Main Hwy, Miami, FL 33133, USA
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Krish Hirani
- Young Leaders Advocacy Group, Diabetes Research Institute Foundation, Hollywood, FL 33021, USA; (B.S.); (K.H.); (M.B.); (J.R.N.L.)
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- American Heritage School, 12200 W Broward Blvd, Plantation, FL 33325, USA
| | - Mateo Blaschke
- Young Leaders Advocacy Group, Diabetes Research Institute Foundation, Hollywood, FL 33021, USA; (B.S.); (K.H.); (M.B.); (J.R.N.L.)
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Coral Gables High School, 450 Bird Rd, Coral Gables, FL 33146, USA
| | - Joana R. N. Lemos
- Young Leaders Advocacy Group, Diabetes Research Institute Foundation, Hollywood, FL 33021, USA; (B.S.); (K.H.); (M.B.); (J.R.N.L.)
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Rahul Mittal
- Young Leaders Advocacy Group, Diabetes Research Institute Foundation, Hollywood, FL 33021, USA; (B.S.); (K.H.); (M.B.); (J.R.N.L.)
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
10
|
Lai Y, Huang X, Sun H, Hui Q, Hu S. Research Progress in the Relationship between Intestinal Flora and Diabetes Mellitus. Endocr Metab Immune Disord Drug Targets 2025; 25:281-290. [PMID: 38956918 DOI: 10.2174/0118715303308965240624054156] [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: 02/06/2024] [Revised: 05/24/2024] [Accepted: 06/04/2024] [Indexed: 07/04/2024]
Abstract
Diabetes mellitus is a common chronic metabolic disease characterized by a high incidence and disability rate. Intestinal flora refers to the microbial community that lives in the intestines and plays a crucial role in maintaining intestinal health and the human immune system. In recent years, an increasing body of research has revealed a close relationship between intestinal flora and diabetes. The pathophysiological mechanisms between them have also been constantly uncovered, and the regulation of intestinal flora has shown promising efficacy in the adjuvant treatment of diabetes. This study mainly summarized the characteristics and mechanisms of intestinal flora in patients with diabetes in recent years, as well as the methods of regulating intestinal flora to prevent and treat diabetes, and prospected the future research direction. This will offer a theoretical basis for the clinical adjuvant treatment of diabetes with intestinal flora and the development of new drugs.
Collapse
Affiliation(s)
- Yingji Lai
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xianfeng Huang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hongwei Sun
- Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qi Hui
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shanshan Hu
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
11
|
Chen K, Wang H, Yang X, Tang C, Hu G, Gao Z. Targeting gut microbiota as a therapeutic target in T2DM: A review of multi-target interactions of probiotics, prebiotics, postbiotics, and synbiotics with the intestinal barrier. Pharmacol Res 2024; 210:107483. [PMID: 39521027 DOI: 10.1016/j.phrs.2024.107483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/11/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
The global epidemic of type 2 diabetes mellitus (T2DM) imposes a substantial burden on public health and healthcare expenditures, thereby driving the pursuit of cost-effective preventive and therapeutic strategies. Emerging evidence suggests a potential association between dysbiosis of gut microbiota and its metabolites with T2DM, indicating that targeted interventions aimed at modulating gut microbiota may represent a promising therapeutic approach for the management of T2DM. In this review, we concentrated on the multifaceted interactions between the gut microbiota and the intestinal barrier in the context of T2DM. We systematically summarized that the imbalance of beneficial gut microbiota and its metabolites may constitute a viable therapeutic approach for the management of T2DM. Meanwhile, the mechanisms by which gut microbiota interventions, such as probiotics, prebiotics, postbiotics, and synbiotics, synergistically improve insulin resistance in T2DM are summarized. These mechanisms include the restoration of gut microbiota structure, upregulation of intestinal epithelial cell proliferation and differentiation, enhancement of tight junction protein expression, promotion of mucin secretion by goblet cells, and the immunosuppressive functions of regulatory T cells (Treg) and M2 macrophages. Collectively, these actions contribute to the amelioration of the body's metabolic inflammatory status. Our objective is to furnish evidence that supports the clinical application of probiotics, prebiotics, and postbiotics in the management of T2DM.
Collapse
Affiliation(s)
- Keyu Chen
- Institute of Metabolic Diseases, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Department of Endocrinology, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Han Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiaofei Yang
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Cheng Tang
- National Key Laboratory of Efficacy and Mechanism on Chinese Medicine for Metabolic Diseases, Beijing Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Guojie Hu
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| | - Zezheng Gao
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| |
Collapse
|
12
|
Dong W, Li Q, Chen L, Tang H, Tu K, Luo L, Jiang L, Huang Y. Association between the gut microbiota and diabetic nephropathy: a two-sample Mendelian randomization study. Ren Fail 2024; 46:2357746. [PMID: 38832498 DOI: 10.1080/0886022x.2024.2357746] [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: 03/05/2024] [Accepted: 05/15/2024] [Indexed: 06/05/2024] Open
Abstract
Numerous studies have revealed a correlation between the risk of developing diabetic nephropathy (DN) and the gut microbiota (GM) composition. However, it remains uncertain whether the GM composition causes DN. We aimed to explore any potential causal links between the GM composition and the risk of developing DN. A meta-analysis conducted by the MiBioGen consortium of the largest genome-wide association study (GWAS) provided aggregated data on the GM. DN data were obtained from the IEU database. The inverse-variance weighting (IVW) method was employed as the primary analytical approach. The IVW analysis indicated that genus Dialister (OR = 0.51, 95% CI: 0.34-0.77, p = 0.00118) was protective against DN. In addition, class Gammaproteobacteria (OR = 0.47, 95% CI: 0.27-0.83, p = 0.0096), class Lentisphaeria (OR =0.76, 95% CI: 0.68-0.99, p = 0.04), order Victivallales (OR = 0.76, 95% CI: 0.58-0.99, p = 0.04), and phylum Proteobacteria (OR = 0.53, 95% CI: 0.33-0.85, p = 0.00872) were negatively associated with the risk of developing DN. Genus LachnospiraceaeUCG008 (OR =1.45, 95% CI: 1.08-1.95, p = 0.01), order Bacteroidales (OR = 1.59, 95% CI: 1.02-2.49, p = 0.04), and genus Terrisporobacter (OR = 1.98, 95% CI: 1.14-3.45, p = 0.015) were positively associated with the risk of developing DN. In this study, we established a causal relationship between the genus Dialister and the risk of developing DN. Further trials are required to confirm the protective effects of probiotics on DN and to elucidate the precise protective mechanisms involving genus Dialister and DN.
Collapse
Affiliation(s)
- Wenjie Dong
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Qiuyu Li
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Lei Chen
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Hui Tang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Kun Tu
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Li Luo
- Department of Pharmacy, West China Second Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Longyang Jiang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yilan Huang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
13
|
Stougaard EB, Tougaard NH, Sivalingam S, Hansen CS, Størling J, Hansen TW, Frimodt-Møller M, Steinert RE, Varasteh S, Groop PH, Salmenkari H, Lehto MJ, Persson F, Rossing P. Effects of probiotics and fibers on markers of nephropathy, inflammation, intestinal barrier dysfunction and endothelial dysfunction in individuals with type 1 diabetes and albuminuria. The ProFOS Study. J Diabetes Complications 2024; 38:108892. [PMID: 39471704 DOI: 10.1016/j.jdiacomp.2024.108892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/09/2024] [Accepted: 10/18/2024] [Indexed: 11/01/2024]
Abstract
AIMS To estimate whether a mix of pre- and probiotics would strengthen the gut barrier and protect the kidneys in individuals with type 1 diabetes and albuminuria. METHODS Randomized, placebo-controlled, crossover study. Forty-one participants received synbiotic (pre- and probiotics) mix or placebo for 12 weeks with 6 weeks washout. Primary endpoint was change from baseline to end-of-period in UACR. Secondary endpoints were changes in endothelial glycocalyx thickness, inflammatory and intestinal barrier dysfunction markers, glomerular filtration rate (GFR) and ambulatory systolic blood pressure. RESULTS Thirty-five participants completed the study. Mean age was 58 (SD 10) years, 73 % (n = 30) were male, median UACR was 134 (IQR 63-293) mg/g, estimated GFR was 75 (30) ml/min/1.73m2. There was no significant difference in UACR with a mean relative change (CI 95 %) from baseline to end-of-treatment of -3.0 (-18.4; 15.5) % in the synbiotic group and -12.0 (-29.6; 9.6) % in the placebo group with no significant difference between treatment periods (9.37 (-25.2; 44.0) percentage points; p = 0.60). No significant beneficial difference in the secondary end points was demonstrated. CONCLUSION Twelve weeks treatment with synbiotic mix had no effect on UACR or on any of the secondary endpoints in subjects with type 1 diabetes and albuminuria.
Collapse
Affiliation(s)
| | | | | | | | - Joachim Størling
- Translational Type 1 Diabetes Research, Steno Diabetes Center Copenhagen, Denmark
| | | | | | - Robert E Steinert
- DSM-Firmenich, Health, Nutrition & Care (HNC), Kaiseraugst, Switzerland; Department of Surgery, Division of Visceral and Transplantation Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Soheil Varasteh
- DSM-Firmenich, Health, Nutrition & Care (HNC), Kaiseraugst, Switzerland
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland; Department of Nephrology, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland; Clinical and Molecular Metabolism, Faculty of Medicine Research Programs, University of Helsinki, Helsinki, Finland
| | - Hanne Salmenkari
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland; Clinical and Molecular Metabolism, Faculty of Medicine Research Programs, University of Helsinki, Helsinki, Finland
| | - Markku J Lehto
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland; Department of Nephrology, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland; Clinical and Molecular Metabolism, Faculty of Medicine Research Programs, University of Helsinki, Helsinki, Finland
| | - Frederik Persson
- Complication Research, Steno Diabetes Center Copenhagen, Denmark
| | - Peter Rossing
- Complication Research, Steno Diabetes Center Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Denmark
| |
Collapse
|
14
|
Mick GJ, McCormick KL. The role of GABA in type 1 diabetes. Front Endocrinol (Lausanne) 2024; 15:1453396. [PMID: 39619323 PMCID: PMC11604429 DOI: 10.3389/fendo.2024.1453396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 10/22/2024] [Indexed: 12/13/2024] Open
Abstract
Gamma aminobutyric acid (GABA) is synthesized from glutamate by glutamic decarboxylase (GAD). The entero-pancreatic biology of GABA, which is produced by pancreatic islets, GAD-expressing microbiota, enteric immune cells, or ingested through diet, supports an essential physiologic role of GABA in the health and disease. Outside the central nervous system (CNS), GABA is uniquely concentrated in pancreatic β-cells. They express GAD65, which is a type 1 diabetes (T1D) autoantigen. Glutamate constitutes 10% of the amino acids in dietary protein and is preeminently concentrated in human milk. GABA is enriched in many foods, such as tomato and fermented cheese, and is an over-the-counter supplement. Selected microbiota in the midgut have the enzymatic capacity to produce GABA. Intestinal microbiota interact with gut-associated lymphoid tissue to maintain host defenses and immune tolerance, which are implicated in autoimmune disease. Although GABA is a widely known inhibitory neurotransmitter, oral GABA does not cross the blood brain barrier. Three diabetes-related therapeutic actions are ascribed to GABA, namely, increasing pancreatic β-cell content, attenuating excess glucagon and tamping down T-cell immune destruction. These salutary actions have been observed in numerous rodent diabetes models that usually employed high or near-continuous GABA doses. Clinical studies, to date, have identified positive effects of oral GABA on peripheral blood mononuclear cell cytokine release and plasma glucagon. Going forward, it is reassuring that oral GABA therapy has been well-tolerated and devoid of serious adverse effects.
Collapse
Affiliation(s)
- Gail J. Mick
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, United States
| | | |
Collapse
|
15
|
Matar A, Damianos JA, Jencks KJ, Camilleri M. Intestinal Barrier Impairment, Preservation, and Repair: An Update. Nutrients 2024; 16:3494. [PMID: 39458489 PMCID: PMC11509958 DOI: 10.3390/nu16203494] [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: 09/19/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Our objective was to review published studies of the intestinal barrier and permeability, the deleterious effects of dietary components (particularly fat), the impact of altered intestinal permeability in disease models and human diseases, the role of the microbiome and epigenomics in control of barrier function, and the opportunities to restore normal barrier function with dietary interventions and products of the microbiota. METHODS We conducted a literature review including the following keywords alone or in combination: intestinal barrier, permeability, microbiome, epigenomics, diet, irritable bowel syndrome, inflammatory bowel disease, probiotics. RESULTS Intestinal permeability is modified by a diet including fat, which increases permeability, and nutrients such as fiber, glutamine, zinc, vitamin D, polyphenols, emulsifiers, and anthocyanins, which decrease permeability. There is significant interaction of the microbiome and barrier function, including the inflammatory of luminal/bacterial antigens, and anti-inflammatory effects of commensals or probiotics and their products, including short-chain fatty acids. Epigenomic modification of barrier functions are best illustrated by effects on junction proteins or inflammation. Detailed documentation of the protective effects of diet, probiotics, prebiotics, and microbiota is provided. CONCLUSION intestinal permeability is a critical factor in protection against gastrointestinal diseases and is impacted by nutrients that preserve or heal and repair the barrier and nurture anti-inflammatory effects.
Collapse
Affiliation(s)
| | | | | | - Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA; (A.M.); (J.A.D.)
| |
Collapse
|
16
|
Luppi S, Aldegheri L, Azzalini E, Pacetti E, Barucca Sebastiani G, Fabiani C, Robino A, Comar M. Unravelling the Role of Gut and Oral Microbiota in the Pediatric Population with Type 1 Diabetes Mellitus. Int J Mol Sci 2024; 25:10611. [PMID: 39408940 PMCID: PMC11477131 DOI: 10.3390/ijms251910611] [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: 09/02/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Type 1 Diabetes Mellitus (T1DM) is a chronic autoimmune disease that results in the destruction of pancreatic β cells, leading to hyperglycaemia and the need for lifelong insulin therapy. Although genetic predisposition and environmental factors are considered key contributors to T1DM, the exact causes of the disease remain partially unclear. Recent evidence has focused on the relationship between the gut, the oral cavity, immune regulation, and systemic inflammation. In individuals with T1DM, changes in the gut and oral microbial composition are commonly observed, indicating that dysbiosis may contribute to immune dysregulation. Gut dysbiosis can influence the immune system through increased intestinal permeability, altered production of short chain fatty acids (SCFAs), and interactions with the mucosal immune system, potentially triggering the autoimmune response. Similarly, oral dysbiosis may contribute to the development of systemic inflammation and thus influence the progression of T1DM. A comprehensive understanding of these relationships is essential for the identification of biomarkers for early diagnosis and monitoring, as well as for the development of therapies aimed at restoring microbial balance. This review presents a synthesis of current research on the connection between T1DM and microbiome dysbiosis, with a focus on the gut and oral microbiomes in pediatric populations. It explores potential mechanisms by which microbial dysbiosis contributes to the pathogenesis of T1DM and examines the potential of microbiome-based therapies, including probiotics, prebiotics, synbiotics, and faecal microbiota transplantation (FMT). This complex relationship highlights the need for longitudinal studies to monitor microbiome changes over time, investigate causal relationships between specific microbial species and T1DM, and develop personalised medicine approaches.
Collapse
Affiliation(s)
- Stefania Luppi
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (S.L.); (L.A.); (M.C.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy; (E.A.); (E.P.)
| | - Luana Aldegheri
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (S.L.); (L.A.); (M.C.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy; (E.A.); (E.P.)
| | - Eros Azzalini
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy; (E.A.); (E.P.)
| | - Emanuele Pacetti
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy; (E.A.); (E.P.)
| | - Giulia Barucca Sebastiani
- Medicine of Services Department, Clinical Analysis Laboratory, Azienda Sanitaria Universitaria Giuliano Isontina, 34125 Trieste, Italy; (G.B.S.); (C.F.)
| | - Carolina Fabiani
- Medicine of Services Department, Clinical Analysis Laboratory, Azienda Sanitaria Universitaria Giuliano Isontina, 34125 Trieste, Italy; (G.B.S.); (C.F.)
| | - Antonietta Robino
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (S.L.); (L.A.); (M.C.)
| | - Manola Comar
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (S.L.); (L.A.); (M.C.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy; (E.A.); (E.P.)
| |
Collapse
|
17
|
Acharya B, Tofthagen M, Maciej-Hulme ML, Suissa MR, Karlsson NG. Limited support for a direct connection between prebiotics and intestinal permeability - a systematic review. Glycoconj J 2024; 41:323-342. [PMID: 39287885 PMCID: PMC11522178 DOI: 10.1007/s10719-024-10165-8] [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: 02/07/2024] [Revised: 08/20/2024] [Accepted: 08/29/2024] [Indexed: 09/19/2024]
Abstract
The intestinal barrier is a selective interface between the body´s external and the internal environment. Its layer of epithelial cells is joined together by tight junction proteins. In intestinal permeability (IP), the barrier is compromised, leading to increased translocation of luminal contents such as large molecules, toxins and even microorganisms. Numerous diseases including Inflammatory Bowel Disease (IBD), Coeliac disease (CD), autoimmune disorders, and diabetes are believed to be associated with IP. Dietary interventions, such as prebiotics, may improve the intestinal barrier. Prebiotics are non-digestible food compounds, that promote the growth and activity of beneficial bacteria in the gut. This systematic review assesses the connection between prebiotic usage and IP. PubMed and Trip were used to identify relevant studies conducted between 2010-2023. Only six studies were found, which all varied in the characteristics of the population, study design, and types of prebiotics interventions. Only one study showed a statistically significant effect of prebiotics on IP. Alteration of intestinal barrier function was measured by lactulose/mannitol, chromium-labelled Ethylenediaminetetraacetic acid (51Cr-EDTA), lactulose/rhamnose, and sucralose/erythritol excretion as well as zonulin and glucagon-like peptide 2 levels. Three studies also conducted gut microbiota assessment, and one of them showed statistically significant improvement of the gut microbiome. This study also reported a decrease in zonulin level. The main conclusion from this review is that there is a lack of human studies in this important field. Futhermore, large population studies and using standardized protocols, would be required to properly assess the impact of prebiotic intervention and improvement on IP.
Collapse
Affiliation(s)
- Binayak Acharya
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, St. Olavs Plass, P.O. Box 4, N-0130, Oslo, Norway
| | - Marthe Tofthagen
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, St. Olavs Plass, P.O. Box 4, N-0130, Oslo, Norway
| | - Marissa L Maciej-Hulme
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, St. Olavs Plass, P.O. Box 4, N-0130, Oslo, Norway
| | - Michal Rachel Suissa
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, St. Olavs Plass, P.O. Box 4, N-0130, Oslo, Norway
| | - Niclas G Karlsson
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, St. Olavs Plass, P.O. Box 4, N-0130, Oslo, Norway.
| |
Collapse
|
18
|
Cao R, Gao T, Yue J, Sun G, Yang X. Disordered Gut Microbiome and Alterations in Metabolic Patterns Are Associated With Hypertensive Left Ventricular Hypertrophy. J Am Heart Assoc 2024; 13:e034230. [PMID: 39342506 DOI: 10.1161/jaha.123.034230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 08/23/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Left ventricular hypertrophy (LVH) is most common when driven by hypertension, and it is a strong independent risk factor for adverse cardiovascular events and death. Some animal models support a role for gut microbiota and metabolites in the development of LVH, but cohort studies confirming these findings in populations are lacking. METHODS AND RESULTS We investigated the alterations of gut microbiota and metabolites in 30 patients with hypertension, 30 patients with hypertensive LVH, and 30 matched controls on the basis of 16S rDNA and metabolomic analyses. Thirty stool and 90 serum samples were collected in fasting conditions. ANOVA/Kruskal-Wallis/Pearson's χ2/Fisher's exact test and Bonferroni's correction were used (P<0.0167) for comparison among the 3 groups. A regression analysis and subgroup analysis were performed between gut microbiota and left ventricular mass index (LVMI) and metabolites and LVMI, respectively. Spearman correlation analysis was performed between metabolites and flora and metabolites and LVMI. We observed LVH-enriched Faecalitalea (β=6758.55 [95% CI, 2080.92-11436.18]; P=0.009), Turicibacter (β=8424.76 [95% CI, 2494.05-14355.47]; P=0.01), Ruminococcus torques group (β=840.88 [95% CI, 223.1-1458.67]; P=0.013), and Erysipelotrichaceae UCG-003 (β=856.37 [95% CI, 182.76-1529.98]; P=0.019) were positively correlated with LVMI. A total of 1141 (in sera) and 2657 (in feces) metabolites were identified. There was a sex-specific association between metabolites and LVMI. Significant changes in metabolic pathways in LVH were also observed, especially bile acid and lipid metabolism pathways. CONCLUSIONS Our study demonstrated the disordered gut microbiota and microbial metabolite profiles in LVH. This highlights the roles of gut bacteria and metabolite in this disease and could lead to new intervention, diagnostic, or management paradigms for LVH. REGISTRATION URL: https://www.chictr.org.cn; Unique Identifier: ChiCTR2200055603.
Collapse
Affiliation(s)
- Rong Cao
- Department of Cardiovascular Medicine Research Institute of Hypertension, The Second Affiliated Hospital of Baotou Medical College Baotou Inner Mongolia China
- Department of Cardiovascular Medicine The First Affiliated Hospital of Fujian Medical University Fuzhou Fujian China
| | - Ting Gao
- Geriatric Department Baotou Central Hospital Baotou Inner Mongolia China
| | - Jianwei Yue
- Department of Cardiovascular Medicine Research Institute of Hypertension, The Second Affiliated Hospital of Baotou Medical College Baotou Inner Mongolia China
| | - Gang Sun
- Department of Cardiovascular Medicine Research Institute of Hypertension, The Second Affiliated Hospital of Baotou Medical College Baotou Inner Mongolia China
| | - Xiaomin Yang
- General Medicine Sir Run Run Shaw Hospital, Zhejiang University Zhejiang China
| |
Collapse
|
19
|
Zaongo SD, Zongo AW, Chen Y. Mechanisms underlying the development of type 1 diabetes in ART-treated people living with HIV: an enigmatic puzzle. Front Immunol 2024; 15:1470308. [PMID: 39257582 PMCID: PMC11383789 DOI: 10.3389/fimmu.2024.1470308] [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: 07/25/2024] [Accepted: 08/12/2024] [Indexed: 09/12/2024] Open
Abstract
The immunopathogenesis of HIV infection remains poorly understood. Despite the widespread use of effective modern antiretroviral therapy (ART), people living with HIV (PLWH) are known to develop several comorbidities, including type 1 diabetes (T1DM). However, the etiology and critical mechanisms accounting for the onset of T1DM in the preceding context remain unknown. This article proposes to address this topic in order to provide further understanding and future research directions.
Collapse
Affiliation(s)
- Silvere D Zaongo
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| | - Abel W Zongo
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, China
| | - Yaokai Chen
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| |
Collapse
|
20
|
Fuhri Snethlage CM, de Wit D, Wortelboer K, Rampanelli E, Hanssen NMJ, Nieuwdorp M. Can fecal microbiota transplantations modulate autoimmune responses in type 1 diabetes? Immunol Rev 2024; 325:46-63. [PMID: 38752578 DOI: 10.1111/imr.13345] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease targeting insulin-producing pancreatic beta cells. T1D is a multifactorial disease incorporating genetic and environmental factors. In recent years, the advances in high-throughput sequencing have allowed researchers to elucidate the changes in the gut microbiota taxonomy and functional capacity that accompany T1D development. An increasing number of studies have shown a role of the gut microbiota in mediating immune responses in health and disease, including autoimmunity. Fecal microbiota transplantations (FMT) have been largely used in murine models to prove a causal role of the gut microbiome in disease progression and have been shown to be a safe and effective treatment in inflammatory human diseases. In this review, we summarize and discuss recent research regarding the gut microbiota-host interactions in T1D, the current advancement in therapies for T1D, and the usefulness of FMT studies to explore microbiota-host immunity encounters in murine models and to shape the course of human type 1 diabetes.
Collapse
Affiliation(s)
- Coco M Fuhri Snethlage
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, Amsterdam, The Netherlands
| | - Douwe de Wit
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, Amsterdam, The Netherlands
| | - Koen Wortelboer
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, Amsterdam, The Netherlands
| | - Elena Rampanelli
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity (AII), Amsterdam, The Netherlands
| | - Nordin M J Hanssen
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, Amsterdam, The Netherlands
- Amsterdam Diabeter Center, Amsterdam UMC, Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, Amsterdam, The Netherlands
- Amsterdam Diabeter Center, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Compare D, Sgamato C, Rocco A, Coccoli P, Ambrosio C, Nardone G. The Leaky Gut and Human Diseases: "Can't Fill the Cup if You Don't Plug the Holes First". Dig Dis 2024; 42:548-566. [PMID: 39047703 DOI: 10.1159/000540379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND The gut barrier is a sophisticated and dynamic system that forms the frontline defense between the external environment and the body's internal milieu and includes various structural and functional components engaged not only in digestion and nutrient absorption but also in immune regulation and overall health maintenance. SUMMARY When one or more components of the intestinal barrier lose their structure and escape their function, this may result in a leaky gut. Mounting evidence emphasizes the crucial role of the gut microbiome in preserving the integrity of the gut barrier and provides insights into the pathophysiological implications of conditions related to leaky gut in humans. Assessment of intestinal permeability has evolved from invasive techniques to noninvasive biomarkers, but challenges remain in achieving consensus about the best testing methods and their accuracy. Research on the modulation of gut permeability is just starting, and although no medical guidelines for the treatment of leaky gut syndrome are available, several treatment strategies are under investigation with promising results. KEY MESSAGES This review discusses the composition of the intestinal barrier, the pathophysiology of the leaky gut and its implications on human health, the measurement of intestinal permeability, and the therapeutic strategies to restore gut barrier integrity.
Collapse
Affiliation(s)
- Debora Compare
- Department of Clinical Medicine and Surgery, Gastroenterology, University Federico II of Naples, Naples, Italy
| | - Costantino Sgamato
- Department of Clinical Medicine and Surgery, Gastroenterology, University Federico II of Naples, Naples, Italy
| | - Alba Rocco
- Department of Clinical Medicine and Surgery, Gastroenterology, University Federico II of Naples, Naples, Italy
| | - Pietro Coccoli
- Department of Clinical Medicine and Surgery, Gastroenterology, University Federico II of Naples, Naples, Italy
| | - Carmen Ambrosio
- Department of Clinical Medicine and Surgery, Gastroenterology, University Federico II of Naples, Naples, Italy
| | - Gerardo Nardone
- Department of Clinical Medicine and Surgery, Gastroenterology, University Federico II of Naples, Naples, Italy
| |
Collapse
|
22
|
Liu XW, Li HL, Ma CY, Shi TY, Wang TY, Yan D, Tang H, Lin H, Deng KJ. Predicting the role of the human gut microbiome in type 1 diabetes using machine-learning methods. Brief Funct Genomics 2024; 23:464-474. [PMID: 38376798 DOI: 10.1093/bfgp/elae004] [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: 10/20/2023] [Revised: 01/12/2024] [Accepted: 01/31/2024] [Indexed: 02/21/2024] Open
Abstract
Gut microbes is a crucial factor in the pathogenesis of type 1 diabetes (T1D). However, it is still unclear which gut microbiota are the key factors affecting T1D and their influence on the development and progression of the disease. To fill these knowledge gaps, we constructed a model to find biomarker from gut microbiota in patients with T1D. We first identified microbial markers using Linear discriminant analysis Effect Size (LEfSe) and random forest (RF) methods. Furthermore, by constructing co-occurrence networks for gut microbes in T1D, we aimed to reveal all gut microbial interactions as well as major beneficial and pathogenic bacteria in healthy populations and type 1 diabetic patients. Finally, PICRUST2 was used to predict Kyoto Encyclopedia of Genes and Genomes (KEGG) functional pathways and KO gene levels of microbial markers to investigate the biological role. Our study revealed that 21 identified microbial genera are important biomarker for T1D. Their AUC values are 0.962 and 0.745 on discovery set and validation set. Functional analysis showed that 10 microbial genera were significantly positively associated with D-arginine and D-ornithine metabolism, spliceosome in transcription, steroid hormone biosynthesis and glycosaminoglycan degradation. These genera were significantly negatively correlated with steroid biosynthesis, cyanoamino acid metabolism and drug metabolism. The other 11 genera displayed an inverse correlation. In summary, our research identified a comprehensive set of T1D gut biomarkers with universal applicability and have revealed the biological consequences of alterations in gut microbiota and their interplay. These findings offer significant prospects for individualized management and treatment of T1D.
Collapse
Affiliation(s)
- Xiao-Wei Liu
- School of Life Science and Technology and Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Han-Lin Li
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Cai-Yi Ma
- School of Life Science and Technology and Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Tian-Yu Shi
- School of Life Science and Technology and Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Tian-Yu Wang
- School of Life Science and Technology and Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Dan Yan
- Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
- Beijing Institute of Clinical Pharmacy, Beijing 100050, China
| | - Hua Tang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
- Basic Medicine Research Innovation Center for Cardiometabolic diseases, Ministry of Education, Luzhou 646000, China
- Central Nervous System Drug Key Laboratory of Sichuan Province, Luzhou 646000, China
| | - Hao Lin
- School of Life Science and Technology and Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Ke-Jun Deng
- School of Life Science and Technology and Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| |
Collapse
|
23
|
Singh J, Vanlallawmzuali, Singh A, Biswal S, Zomuansangi R, Lalbiaktluangi C, Singh BP, Singh PK, Vellingiri B, Iyer M, Ram H, Udey B, Yadav MK. Microbiota-brain axis: Exploring the role of gut microbiota in psychiatric disorders - A comprehensive review. Asian J Psychiatr 2024; 97:104068. [PMID: 38776563 DOI: 10.1016/j.ajp.2024.104068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/28/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024]
Abstract
Mental illness is a hidden epidemic in modern science that has gradually spread worldwide. According to estimates from the World Health Organization (WHO), approximately 10% of the world's population suffers from various mental diseases each year. Worldwide, financial and health burdens on society are increasing annually. Therefore, understanding the different factors that can influence mental illness is required to formulate novel and effective treatments and interventions to combat mental illness. Gut microbiota, consisting of diverse microbial communities residing in the gastrointestinal tract, exert profound effects on the central nervous system through the gut-brain axis. The gut-brain axis serves as a conduit for bidirectional communication between the two systems, enabling the gut microbiota to affect emotional and cognitive functions. Dysbiosis, or an imbalance in the gut microbiota, is associated with an increased susceptibility to mental health disorders and psychiatric illnesses. Gut microbiota is one of the most diverse and abundant groups of microbes that have been found to interact with the central nervous system and play important physiological functions in the human gut, thus greatly affecting the development of mental illnesses. The interaction between gut microbiota and mental health-related illnesses is a multifaceted and promising field of study. This review explores the mechanisms by which gut microbiota influences mental health, encompassing the modulation of neurotransmitter production, neuroinflammation, and integrity of the gut barrier. In addition, it emphasizes a thorough understanding of how the gut microbiome affects various psychiatric conditions.
Collapse
Affiliation(s)
- Jawahar Singh
- Department of Psychiatry, All India Institute of Medical Sciences (AIIMS), Bathinda, Punjab, India
| | - Vanlallawmzuali
- Department of Biotechnology, Mizoram Central University, Pachhunga University College Campus, Aizawl, Mizoram, India
| | - Amit Singh
- Department of Microbiology Central University of Punjab, Bathinda 151401, India
| | - Suryanarayan Biswal
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda 151401, India
| | - Ruth Zomuansangi
- Department of Microbiology Central University of Punjab, Bathinda 151401, India
| | - C Lalbiaktluangi
- Department of Microbiology Central University of Punjab, Bathinda 151401, India
| | - Bhim Pratap Singh
- Department of Agriculture and Environmental Sciences (AES), National Institute of Food Technology Entrepreneurship and Management (NIFTEM), Sonepat, Haryana, India
| | - Prashant Kumar Singh
- Department of Biotechnology, Pachhunga University College Campus, Mizoram University (A Central University), Aizawl 796001, Mizoram, India
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda, Punjab 151401, India
| | - Mahalaxmi Iyer
- Department of Microbiology Central University of Punjab, Bathinda 151401, India
| | - Heera Ram
- Department of Zoology, Jai Narain Vyas University, Jodhpur, Rajasthan 342001, India
| | - Bharat Udey
- Department of Psychiatry, All India Institute of Medical Sciences (AIIMS), Bathinda, Punjab, India
| | - Mukesh Kumar Yadav
- Department of Microbiology Central University of Punjab, Bathinda 151401, India.
| |
Collapse
|
24
|
Basuray N, Deehan EC, Vieira FT, Avedzi HM, Duke RL, Colín-Ramírez E, Tun HM, Zhang Z, Wine E, Madsen KL, Field CJ, Haqq AM. Dichotomous effect of dietary fiber in pediatrics: a narrative review of the health benefits and tolerance of fiber. Eur J Clin Nutr 2024; 78:557-568. [PMID: 38480843 DOI: 10.1038/s41430-024-01429-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 02/27/2024] [Accepted: 03/01/2024] [Indexed: 03/18/2024]
Abstract
Dietary fibers are associated with favorable gastrointestinal, immune, and metabolic health outcomes when consumed at sufficient levels. Despite the well-described benefits of dietary fibers, children and adolescents continue to fall short of daily recommended levels. This gap in fiber intake (i.e., "fiber gap") might increase the risk of developing early-onset pediatric obesity and obesity-related comorbidities such as type 2 diabetes mellitus into adulthood. The structure-dependent physicochemical properties of dietary fiber are diverse. Differences in solubility, viscosity, water-holding capacity, binding capability, bulking effect, and fermentability influence the physiological effects of dietary fibers that aid in regulating appetite, glycemic and lipidemic responses, and inflammation. Of growing interest is the fermentation of fibers by the gut microbiota, which yields both beneficial and less favorable end-products such as short-chain fatty acids (e.g., acetate, propionate, and butyrate) that impart metabolic and immunomodulatory properties, and gases (e.g., hydrogen, carbon dioxide, and methane) that cause gastrointestinal symptoms, respectively. This narrative review summarizes (1) the implications of fibers on the gut microbiota and the pathophysiology of pediatric obesity, (2) some factors that potentially contribute to the fiber gap with an emphasis on undesirable gastrointestinal symptoms, (3) some methods to alleviate fiber-induced symptoms, and (4) the therapeutic potential of whole foods and commonly marketed fiber supplements for improved health in pediatric obesity.
Collapse
Affiliation(s)
- Nandini Basuray
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Edward C Deehan
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE, USA
- Nebraska Food for Health Center, Lincoln, NE, USA
| | - Flávio T Vieira
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Hayford M Avedzi
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Reena L Duke
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | | | - Hein M Tun
- JC School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Zhengxiao Zhang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, China
| | - Eytan Wine
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Karen L Madsen
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Andrea M Haqq
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada.
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
25
|
Zhang Y, Huang A, Li J, Munthali W, Cao S, Putri UMP, Yang L. The Effect of Microbiome-Modulating Agents (MMAs) on Type 1 Diabetes: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Nutrients 2024; 16:1675. [PMID: 38892608 PMCID: PMC11174426 DOI: 10.3390/nu16111675] [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: 04/30/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Gut microbiome-modulating agents (MMAs), including probiotics, prebiotics, postbiotics, and synbiotics, are shown to ameliorate type 1 diabetes (T1D) by restoring the microbiome from dysbiosis. The objective of this systematic review and meta-analysis was to assess the impact of MMAs on hemoglobin A1c (HbA1c) and biomarkers associated with (T1D). A comprehensive search was conducted in PubMed, Web of Science, Embase, Cochrane Library, National Knowledge Infrastructure, WeiPu, and WanFang Data up to 30 November 2023. Ten randomized controlled trials (n = 630) were included, with study quality evaluated using the Cochrane risk-of-bias tool. Random-effect models with standardized mean differences (SMDs) were utilized. MMA supplementation was associated with improvements in HbA1c (SMD = -0.52, 95% CI [-0.83, -0.20]), daily insulin usage (SMD = -0.41, 95% confidence interval (CI) [-0.76, -0.07]), and fasting C-peptide (SMD = 0.99, 95% CI [0.17, 1.81]) but had no effects on FBG, CRP, TNF-α, IL-10, LDL, HDL, and the Shannon index. Subgroup analysis of HbA1c indicated that a long-term intervention (>3 months) might exert a more substantial effect. These findings suggest an association between MMAs and glycemic control in T1D. Further large-scale clinical trials are necessary to confirm these findings with investigations on inflammation and gut microbiota composition while adjusting confounding factors such as diet, physical activity, and the dose and form of MMA intervention.
Collapse
Affiliation(s)
- Ying Zhang
- Xiangya School of Public Health, Central South University, Changsha 410128, China; (Y.Z.)
| | - Aiying Huang
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Jun Li
- School of Psychology, South China Normal University, Guangzhou 510631, China
| | - William Munthali
- Xiangya School of Public Health, Central South University, Changsha 410128, China; (Y.Z.)
| | - Saiying Cao
- Xiangya School of Public Health, Central South University, Changsha 410128, China; (Y.Z.)
| | | | - Lina Yang
- Xiangya School of Public Health, Central South University, Changsha 410128, China; (Y.Z.)
| |
Collapse
|
26
|
Baghel K, Khan A, Kango N. Role of Synbiotics (Prebiotics and Probiotics) as Dietary Supplements in Type 2 Diabetes Mellitus Induced Health Complications. J Diet Suppl 2024; 21:677-708. [PMID: 38622882 DOI: 10.1080/19390211.2024.2340509] [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] [Indexed: 04/17/2024]
Abstract
Diabetes is a metabolic disorder whose prevalence has become a worrying condition in recent decades. Chronic diabetes can result in serious health conditions such as impaired kidney function, stroke, blindness, and myocardial infarction. Despite a variety of currently available treatments, cases of diabetes and its complications are on the rise. This review article provides a comprehensive account of the ameliorative effect of prebiotics and probiotics individually or in combination i.e. synbiotics on health complications induced by Type 2 Diabetes Mellitus (T2DM). Recent advances in the field underscore encouraging outcomes suggesting the consumption of synbiotics leads to favorable changes in the gut microbiota. These changes result in the production of bioactive metabolites such as short-chain fatty acids (crucial for lowering blood sugar levels), reducing inflammation, preventing insulin resistance, and encouraging the release of glucagon-like peptide-1 in the host. Notably, novel strategies supplementing synbiotics to support gut microbiota are gaining attraction as pivotal interventions in mitigating T2DM-induced health complications. Thus, by nurturing a symbiotic relationship between prebiotics and probiotics i.e. synbiotics, these interventions hold promise in reshaping the microbial landscape of the gut thereby offering a multifaceted approach to managing T2DM and its associated morbidities. Supporting the potential of synbiotics underscores a paradigm shift toward holistic and targeted interventions in diabetes management, offering prospects for improved outcomes and enhanced quality of life for affected individuals. Nevertheless, more research needs to be done to better understand the single and multispecies pre/pro and synbiotics in the prevention and management of T2DM-induced health complications.
Collapse
Affiliation(s)
- Kalpana Baghel
- Department of Microbiology, School of Biological Sciences, Dr Harisingh Gour Vishwavidyalaya (A Central University), Sagar, MP, India
- Department of Zoology, School of Biological Sciences, Dr Harisingh Gour Vishwavidyalaya (A Central University), Sagar, MP, India
| | - Aamir Khan
- Department of Zoology, School of Biological Sciences, Dr Harisingh Gour Vishwavidyalaya (A Central University), Sagar, MP, India
| | - Naveen Kango
- Department of Microbiology, School of Biological Sciences, Dr Harisingh Gour Vishwavidyalaya (A Central University), Sagar, MP, India
| |
Collapse
|
27
|
Kaźmierczak-Siedlecka K, Maciejewska-Markiewicz D, Sykulski M, Gruszczyńska A, Herman-Iżycka J, Wyleżoł M, Katarzyna Petriczko K, Palma J, Jakubczyk K, Janda-Milczarek K, Skonieczna-Żydecka K, Stachowska E. Gut Microbiome-How Does Two-Month Consumption of Fiber-Enriched Rolls Change Microbiome in Patients Suffering from MASLD? Nutrients 2024; 16:1173. [PMID: 38674864 PMCID: PMC11053994 DOI: 10.3390/nu16081173] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/04/2024] [Accepted: 04/07/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND The occurrence of metabolic dysfunction-associated steatotic liver disease (MASLD) is a growing global problem which commonly affects patients with co-existing diseases/conditions, such as type 2 diabetes and dyslipidemia. The effective treatment of MASLD is still limited; however, diet plays a significant role in its management. There are multiple beneficial properties of dietary fiber, including its ability to modify the gut microbiome. Therefore, the aim of this study was to determine the effect of the consumption of fiber-enriched rolls on the gut microbiome and microbial metabolites in patients suffering from MASLD. METHODS The participants were recruited according to the inclusion criteria and were required to consume fiber-enriched rolls containing either 6 g or 12 g of fiber. There were three assessment timepoints, when the anthropometric and laboratory parameters were measured, and 16s on nanopore sequencing of the fecal microbiome was conducted. RESULTS Firmicutes and Bacteroidetes were the most abundant phyla in the patients living with MASLD. It was demonstrated that the amount of short-chain fatty acids (SCFAs) changed after the consumption of fiber-enriched rolls; however, this was strongly associated with both the timepoint and the type of SCFAs-acetate and butyrate. Additionally, the high-fiber diet was related to the increase in phyla diversity (p = 0.006571). CONCLUSIONS Overall, the introduction of an appropriate amount of fiber to the diet seems to be promising for patients suffering from MASLD due to its ability to create an improvement in gut microbiome-related aspects.
Collapse
Affiliation(s)
- Karolina Kaźmierczak-Siedlecka
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-211 Gdansk, Poland;
| | - Dominika Maciejewska-Markiewicz
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (D.M.-M.); (K.J.); (K.J.-M.)
| | - Maciej Sykulski
- Department of Medical Genetics, Medical University of Warsaw, 02-106 Warsaw, Poland
| | - Agata Gruszczyńska
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | | | - Mariusz Wyleżoł
- Department of General, Vascular and Oncological Surgery, Medical University of Warsaw, 02-091 Warsaw, Poland;
| | - Karolina Katarzyna Petriczko
- Translational Medicine Group, Pomeranian Medical University, 70-204 Szczecin, Poland;
- Department of Gastroenterology and Internal Medicine, SPWSZ Hospital, 71-455 Szczecin, Poland
| | - Joanna Palma
- Department of Biochemical Sciences, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460 Szczecin, Poland;
| | - Karolina Jakubczyk
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (D.M.-M.); (K.J.); (K.J.-M.)
| | - Katarzyna Janda-Milczarek
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (D.M.-M.); (K.J.); (K.J.-M.)
| | - Karolina Skonieczna-Żydecka
- Department of Biochemical Sciences, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460 Szczecin, Poland;
| | - Ewa Stachowska
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (D.M.-M.); (K.J.); (K.J.-M.)
| |
Collapse
|
28
|
Ribeiro MC, Levi Y, Moraschini V, Messora MR, Furlaneto FAC. Effects of Prebiotic Therapy on Gastrointestinal Microbiome of Individuals with Different Inflammatory Conditions: A Systematic Review of Randomized Controlled Trials. Probiotics Antimicrob Proteins 2024; 16:673-695. [PMID: 37093515 DOI: 10.1007/s12602-023-10075-5] [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] [Accepted: 04/06/2023] [Indexed: 04/25/2023]
Abstract
Prebiotics are substrates selectively utilized by host microorganisms conferring a health benefit. The effects of prebiotics on the gut microbiome of individuals with inflammatory processes need further investigations. The purpose of this study was to evaluate the effects of prebiotics on the gastrointestinal microbiome of individuals with some types of inflammatory conditions. Randomized controlled clinical trials (RCTs) evaluating the effects of different prebiotics on the gut microbiome were included. A systematic review of the literature including searches in PubMed/MEDLINE, EMBASE, Cochrane Library, Web of Science, and Scopus databases was performed until 23 March 2023. The risk of bias was assessed using the Cochrane Collaboration's criteria. Qualitative data was tabulated to facilitate comparisons and represented in the form of descriptive statistics and summary tables. Thirty trials, ranging from 12 to 135 patients, were included. The most commonly used prebiotic type was inulin-type fructans, and the treatment duration ranged from 1 to 36 weeks. The majority of the trials investigated the gut microbiome using 16 s rRNA gene sequencing on the Illumina Miseq platform. In general, prebiotic therapy exerted positive effects on inflammatory conditions. An increase in Bifidobacterium genus was the most common shift in bacterial composition observed. Within the limits of this systematic review, it can be suggested that prebiotic therapy presents the potential to favorably modulate the gastrointestinal microbiome of individuals with different types of inflammatory conditions.
Collapse
Affiliation(s)
- M C Ribeiro
- Department of Oral Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of Sao Paulo - USP, Av. Café S/N, 14020-150, Ribeirao Preto, São Paulo, Brazil
| | - Ylas Levi
- Department of Oral Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of Sao Paulo - USP, Av. Café S/N, 14020-150, Ribeirao Preto, São Paulo, Brazil
| | - V Moraschini
- Department of Periodontology, Dental Research Division, School of Dentistry, Veiga de Almeida University, Rio de Janeiro, Brazil
| | - M R Messora
- Department of Oral Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of Sao Paulo - USP, Av. Café S/N, 14020-150, Ribeirao Preto, São Paulo, Brazil
| | - F A C Furlaneto
- Department of Oral Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of Sao Paulo - USP, Av. Café S/N, 14020-150, Ribeirao Preto, São Paulo, Brazil.
| |
Collapse
|
29
|
Guimarães JB, Rodrigues VF, Pereira ÍS, Manso GMDC, Elias-Oliveira J, Leite JA, Waldetario MCGM, de Oliveira S, Gomes ABDSP, Faria AMC, Ramos SG, Bonato VLD, Silva JS, Vinolo MAR, Sampaio UM, Clerici MTPS, Carlos D. Inulin prebiotic ameliorates type 1 diabetes dictating regulatory T cell homing via CCR4 to pancreatic islets and butyrogenic gut microbiota in murine model. J Leukoc Biol 2024; 115:483-496. [PMID: 37947010 DOI: 10.1093/jleuko/qiad132] [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: 11/29/2022] [Revised: 09/04/2023] [Accepted: 10/14/2023] [Indexed: 11/12/2023] Open
Abstract
Gut dysbiosis is linked to type 1 diabetes mellitus (T1D). Inulin (INU), a prebiotic, modulates the gut microbiota, promoting beneficial bacteria that produce essential short-chain fatty acids for immune regulation. However, how INU affects T1D remains uncertain. Using a streptozotocin-induced (STZ) mouse model, we studied INU's protective effects. Remarkably, STZ + INU mice resisted T1D, with none developing the disease. They had lower blood glucose, reduced pancreatic inflammation, and normalized serum insulin compared with STZ + SD mice. STZ + INU mice also had enhanced mucus production, abundant Bifidobacterium, Clostridium cluster IV, Akkermansia muciniphila, and increased fecal butyrate. In cecal lymph nodes, we observed fewer CD4+Foxp3+ regulatory T cells expressing CCR4 and more Foxp3+CCR4+ cells in pancreatic islets, with higher CCL17 expression. This phenotype was absent in CCR4-deficient mice on INU. INU supplementation effectively protects against experimental T1D by recruiting CCR4+ regulatory T cells via CCL17 into the pancreas and altering the butyrate-producing microbiota.
Collapse
Affiliation(s)
- Jhefferson Barbosa Guimarães
- Laboratory of Imunorregulation of Metabolic Diseases, Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ave. Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Vanessa Fernandes Rodrigues
- Laboratory of Imunorregulation of Metabolic Diseases, Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ave. Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Ítalo Sousa Pereira
- Laboratory of Imunorregulation of Metabolic Diseases, Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ave. Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Gabriel Martins da Costa Manso
- Laboratory of Imunorregulation of Metabolic Diseases, Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ave. Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Jefferson Elias-Oliveira
- Laboratory of Imunorregulation of Metabolic Diseases, Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ave. Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Jefferson Antônio Leite
- Laboratory of Imunorregulation of Metabolic Diseases, Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ave. Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | | | - Sarah de Oliveira
- Laboratory of Immunoinflammation, Department of Genetics and Evolution, Microbiology and Immunology, Institute of Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Arilson Bernardo Dos Santos Pereira Gomes
- Laboratory of Immunoinflammation, Department of Genetics and Evolution, Microbiology and Immunology, Institute of Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Ana Maria Caetano Faria
- Department of Biochemistry and Immunology, Institute of Biological Sciences, University of Minas Gerais, Belo Horizonte, Minas Gerais,31270-901, Brazil
| | - Simone Gusmão Ramos
- Laboratory of Pathology, Department of Pathology, Ribeirão Preto Medical School, University of São Paulo, Ave. Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Vânia L D Bonato
- Laboratory of Immunology and Pulmonary Inflammation, Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Ave. Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - João Santana Silva
- Fiocruz-Bi-Institutional Translational Medicine Plataform, University of São Paulo, Ave. Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Marco Aurélio Ramirez Vinolo
- Laboratory of Immunoinflammation, Department of Genetics and Evolution, Microbiology and Immunology, Institute of Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Ulliana Marques Sampaio
- Department of Food Science and Nutrition, School of Food Engineering, State University of Campinas, Campinas, São Paulo, 13083-970, Brazil
| | - Maria Teresa Pedrosa Silva Clerici
- Department of Food Science and Nutrition, School of Food Engineering, State University of Campinas, Campinas, São Paulo, 13083-970, Brazil
| | - Daniela Carlos
- Laboratory of Imunorregulation of Metabolic Diseases, Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ave. Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil
| |
Collapse
|
30
|
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
|
31
|
Li Y, Liu Q, Zhang L, Zou J, He R, Zhou Y, Qian C, Zhu Y, Chen R, Zhang Y, Cai P, Wang M, Shao W, Ji M, Wu H, Zhang F, Liu Z, Liu Y. Washed microbiota transplantation reduces glycemic variability in unstable diabetes. J Diabetes 2024; 16:e13485. [PMID: 37846600 PMCID: PMC10859319 DOI: 10.1111/1753-0407.13485] [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: 12/08/2022] [Revised: 09/17/2023] [Accepted: 09/25/2023] [Indexed: 10/18/2023] Open
Abstract
BACKGROUND Dysbiosis of gut microbiota is causally linked to impaired host glucose metabolism. We aimed to study effects of the new method of fecal microbiota transplantation, washed microbiota transplantation (WMT), on reducing glycemic variability (GV) in unstable diabetes. METHODS Fourteen eligible patients received three allogenic WMTs and were followed up at 1 week, 1 month, and 3 months. Primary outcomes were daily insulin dose, glucose excursions during meal tests, and GV indices calculated from continuous monitoring or self-monitoring glucose values. Secondary outcomes were multiomics data, including 16S rRNA gene sequencing, metagenomics, and metabolomics to explore underlying mechanisms. RESULTS Daily insulin dose and glucose excursions markedly dropped, whereas GV indices significantly improved up to 1 month. WMT increased gut microbial alpha diversity, beta diversity, and network complexity. Taxonomic changes featured lower abundance of genera Bacteroides and Escherichia-Shigella, and higher abundance of genus Prevotella. Metagenomics functional annotations revealed enrichment of distinct microbial metabolic pathways, including methane biosynthesis, citrate cycle, amino acid degradation, and butyrate production. Derived metabolites correlated significantly with improved GV indices. WMT did not change circulating inflammatory cytokines, enteroendocrine hormones, or C-peptide. CONCLUSIONS WMT showed strong ameliorating effect on GV, raising the possibility of targeting gut microbiota as an effective regimen to reduce GV in diabetes.
Collapse
Affiliation(s)
- Yangyang Li
- Department of Endocrinology, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Qing Liu
- Department of Endocrinology, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Lingyu Zhang
- Department of Endocrinology, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Jing Zou
- Department of Endocrinology, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Rongbo He
- Department of Endocrinology, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Ying Zhou
- Department of Endocrinology, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Chen Qian
- Department of Endocrinology, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Yuxiao Zhu
- Department of Endocrinology, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Rourou Chen
- Department of Endocrinology, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Ying Zhang
- Department of Endocrinology, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Pengpeng Cai
- Digestive Endoscopy Center, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Miao Wang
- Division of Microbiotherapy, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Wei Shao
- Department of Science and Technology, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Minjun Ji
- Department of Pathogen Biology, Jiangsu Province Key Laboratory of Modern Pathogen BiologyNanjing Medical UniversityNanjingChina
| | - Hao Wu
- Human Phenome InstituteFudan UniversityShanghaiChina
| | - Faming Zhang
- Division of Microbiotherapy, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
- Medical Center for Digestive Diseasesthe Second Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Key Lab of Holistic Integrative EnterologyNanjing Medical UniversityNanjingChina
| | - Zejian Liu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular BiologyNanjing Medical UniversityNanjingChina
| | - Yu Liu
- Department of Endocrinology, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| |
Collapse
|
32
|
Ye XY, Chen JY, Wu LH, Luo DP, Ye XH, Wu LQ, He XX. Washed microbiota transplantation improves symptoms and intestinal barrier function in patients with functional bowel disorders: a propensity-score matching analysis. BMC Gastroenterol 2024; 24:45. [PMID: 38262980 PMCID: PMC10804514 DOI: 10.1186/s12876-024-03131-z] [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: 03/28/2023] [Accepted: 01/09/2024] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND Alterations in the intestinal microbiota may play a role in the pathogenesis of functional bowel disorders (FBDs). Probiotics are widely used to improve intestinal dysbacteriosis in FBDs. In the context of FBDs, washed microbiota transplantation (WMT) appear to be a promising therapeutic option. We aimed to compare probiotics with WMT by using a propensity-score matching analysis (PSMA). METHODS We conducted a retrospective investigation of 103 patients with FBDs, including irritable bowel syndrome (IBS), functional constipation (FC), functional diarrhea (FDr), functional abdominal bloating (FAB). Patients were divided into the WMT group or probiotics group (taking probiotics capsules). Data on the following parameters were matched for PSMA: age; sex; disease course; body mass index; anxiety; insomnia; tobacco smoking; alcohol consumption; and levels of D-lactate, diamine oxidase, and lipopolysaccharide. Intestinal barrier function (IBF) and symptoms were evaluated both before and after treatment initiation. Prognostic factors were assessed by Cox proportional hazards regression analysis. RESULTS PSMA identified in 34 matched pairs (11 IBS, 12 FC, 7 FDr, and 4 FAB in the probiotics group and 14 IBS, 13 FC, 5 FDr, and 2 FAB in the WMT group. Improvement of FBD symptoms was greater with WMT than probiotics (P = 0.002). The WMT group had significantly fewer patients with intestinal barrier damage than the probiotics group (38.2% vs. 67.6%, P = 0.041). This improvement of FBD with WMT was further reflected as a reduction in D-lactate levels (P = 0.031). Increased D-lactate levels which were identified as a prognostic factor for FBDs (HR = 0.248, 95%CI 0.093-0.666, P = 0.006) in multivariate Cox regression analysis. CONCLUSION WMT could improve symptoms and IBF in patients with FBDs. Increased D-lactate levels in patients with FBDs may predict a favorable response to WMT treatment.
Collapse
Affiliation(s)
- Xiao-Yan Ye
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, 19 Nonglinxia Road, Yuexiu District, 510030, Guangzhou, Guangdong Province, China
- Department of Gastroenterology, Research Center for Engineering Techniques of Microbiota -Targeted Therapies of Guangdong Province, 510030, Guangzhou, Guangdong Province, China
| | - Jun-Yi Chen
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, 19 Nonglinxia Road, Yuexiu District, 510030, Guangzhou, Guangdong Province, China
- Department of Gastroenterology, Research Center for Engineering Techniques of Microbiota -Targeted Therapies of Guangdong Province, 510030, Guangzhou, Guangdong Province, China
| | - Li-Hao Wu
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, 19 Nonglinxia Road, Yuexiu District, 510030, Guangzhou, Guangdong Province, China
- Department of Gastroenterology, Research Center for Engineering Techniques of Microbiota -Targeted Therapies of Guangdong Province, 510030, Guangzhou, Guangdong Province, China
| | - Dan-Ping Luo
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, 19 Nonglinxia Road, Yuexiu District, 510030, Guangzhou, Guangdong Province, China
- Department of Gastroenterology, Research Center for Engineering Techniques of Microbiota -Targeted Therapies of Guangdong Province, 510030, Guangzhou, Guangdong Province, China
| | - Xiao-Huo Ye
- Department of Pharmacy, Heyuan Health School, 517000, Heyuan, Guangdong Province, China
| | - Li-Quan Wu
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, 19 Nonglinxia Road, Yuexiu District, 510030, Guangzhou, Guangdong Province, China
- Department of Gastroenterology, Research Center for Engineering Techniques of Microbiota -Targeted Therapies of Guangdong Province, 510030, Guangzhou, Guangdong Province, China
| | - Xing-Xiang He
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, 19 Nonglinxia Road, Yuexiu District, 510030, Guangzhou, Guangdong Province, China.
- Department of Gastroenterology, Research Center for Engineering Techniques of Microbiota -Targeted Therapies of Guangdong Province, 510030, Guangzhou, Guangdong Province, China.
| |
Collapse
|
33
|
Vandenbempt V, Eski SE, Brahma MK, Li A, Negueruela J, Bruggeman Y, Demine S, Xiao P, Cardozo AK, Baeyens N, Martelotto LG, Singh SP, Mariño E, Gysemans C, Gurzov EN. HAMSAB diet ameliorates dysfunctional signaling in pancreatic islets in autoimmune diabetes. iScience 2024; 27:108694. [PMID: 38213620 PMCID: PMC10783594 DOI: 10.1016/j.isci.2023.108694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 10/23/2023] [Accepted: 12/05/2023] [Indexed: 01/13/2024] Open
Abstract
An altered gut microbiota is associated with type 1 diabetes (T1D), affecting the production of short-chain fatty acids (SCFA) and glucose homeostasis. We previously demonstrated that enhancing serum acetate and butyrate using a dietary supplement (HAMSAB) improved glycemia in non-obese diabetic (NOD) mice and patients with established T1D. The effects of SCFA on immune-infiltrated islet cells remain to be clarified. Here, we performed single-cell RNA sequencing on islet cells from NOD mice fed an HAMSAB or control diet. HAMSAB induced a regulatory gene expression profile in pancreas-infiltrated immune cells. Moreover, HAMSAB maintained the expression of β-cell functional genes and decreased cellular stress. HAMSAB-fed mice showed preserved pancreatic endocrine cell identity, evaluated by decreased numbers of poly-hormonal cells. Finally, SCFA increased insulin levels in human β-like cells and improved transplantation outcome in NOD/SCID mice. Our findings support the use of metabolite-based diet as attractive approach to improve glucose control in T1D.
Collapse
Affiliation(s)
- Valerie Vandenbempt
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, 1070 Brussels, Belgium
| | - Sema Elif Eski
- IRIBHM, Université libre de Bruxelles, 1070 Brussels, Belgium
| | - Manoja K. Brahma
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, 1070 Brussels, Belgium
| | - Ao Li
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, 1070 Brussels, Belgium
| | - Javier Negueruela
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, 1070 Brussels, Belgium
| | - Ylke Bruggeman
- Clinical and Experimental Endocrinology (CEE), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Campus Gasthuisberg O&N 1, KU Leuven, 3000 Leuven, Belgium
| | - Stéphane Demine
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, 1070 Brussels, Belgium
| | - Peng Xiao
- Inflammatory and Cell Death Signaling in Diabetes group, Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, 1070 Brussels, Belgium
| | - Alessandra K. Cardozo
- Inflammatory and Cell Death Signaling in Diabetes group, Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, 1070 Brussels, Belgium
| | - Nicolas Baeyens
- Laboratoire de Physiologie et de Pharmacologie, Université Libre de Bruxelles, 1000 Brussels, Belgium
| | - Luciano G. Martelotto
- Single Cell and Spatial-Omics Laboratory, Adelaide Centre of Epigenetics, University of Adelaide, Adelaide, SA 5005, Australia
| | | | - Eliana Mariño
- Infection and Immunity Program, Biomedicine Discovery Institute, Department of Biochemistry, Monash University, Melbourne, VIC 3800, Australia
- ImmunoBiota Therapeutics Pty Ltd, Melbourne, VIC 3187, Australia
| | - Conny Gysemans
- Clinical and Experimental Endocrinology (CEE), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Campus Gasthuisberg O&N 1, KU Leuven, 3000 Leuven, Belgium
| | - Esteban N. Gurzov
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, 1070 Brussels, Belgium
- WELBIO Department, WEL Research Institute, Avenue Pasteur 6, 1300 Wavre, Belgium
| |
Collapse
|
34
|
Correa ADC, Lopes MS, Perna RF, Silva EK. Fructan-type prebiotic dietary fibers: Clinical studies reporting health impacts and recent advances in their technological application in bakery, dairy, meat products and beverages. Carbohydr Polym 2024; 323:121396. [PMID: 37940290 DOI: 10.1016/j.carbpol.2023.121396] [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: 06/15/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 11/10/2023]
Abstract
Fructooligosaccharides (FOS) and inulin are the most used fructans in food manufacturing, including bakery, dairy, meat products and beverages. In this context, this review investigated the recent findings concerning health claims associated with a diet supplemented with fructans according to human trial results. Fructans have been applied in different food classes due to their proven benefits to human health. Human clinical trials have revealed several effects of fructans supplementation on health such as improved glycemic control, growth of beneficial gut bacteria, weight management, positive influence on immune function, and others. These dietary fibers have a wide range of compounds with different molecular sizes, implying a great variety of technological properties depending on the food application of interest. Inulin has been mainly applied as a fat substitute and prebiotic ingredient. In general, inulin reduces the energy content and improves the structure, viscosity, emulsion, and water retention parameters of food products. Meanwhile, FOS have been more successful when used as a sucrose substitute and prebiotic ingredient. However, overall, FOS and inulin are promising alternatives for the development of structured systems dedicated to increase the functionality of foods and beverages besides reducing fat in bakery, dairy, and meat products.
Collapse
Affiliation(s)
- Aline de Carvalho Correa
- Graduate Program in Chemical Engineering, Institute of Science and Technology, Federal University of Alfenas - Campus Poços de Caldas, 37715-400 Poços de Caldas, Minas Gerais, Brazil
| | - Melina Savioli Lopes
- Graduate Program in Chemical Engineering, Institute of Science and Technology, Federal University of Alfenas - Campus Poços de Caldas, 37715-400 Poços de Caldas, Minas Gerais, Brazil
| | - Rafael Firmani Perna
- Graduate Program in Chemical Engineering, Institute of Science and Technology, Federal University of Alfenas - Campus Poços de Caldas, 37715-400 Poços de Caldas, Minas Gerais, Brazil
| | - Eric Keven Silva
- School of Food Engineering, University of Campinas, 13083-862, Campinas, São Paulo, Brazil.
| |
Collapse
|
35
|
Chu NHS, He J, Leung KHT, Ma RCW, Lee JYS, Varney J, Chan JCN, Muir JG, Chow E. Higher Short-Chain Fermentable Carbohydrates Are Associated with Lower Body Fat and Higher Insulin Sensitivity in People with Prediabetes. Nutrients 2023; 15:5070. [PMID: 38140329 PMCID: PMC10745595 DOI: 10.3390/nu15245070] [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: 10/17/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
The quality of carbohydrates has metabolic consequences in people with prediabetes. However, the causality of short-chain fermentable carbohydrate intakes and metabolic parameters has not been explored in the prediabetic or diabetic population. We investigated associations between different types of carbohydrates, including fermentable oligosaccharides, disaccharides, monosaccharides, polyols (FODMAPs), and polysaccharides (dietary fibre), and body composition and glucose/insulin responses in subjects with prediabetes. In this prospective cross-sectional study, 177 subjects with impaired glucose tolerance (IGT) (mean age: 60 (54-62) years, 41% men) underwent an assessment of body composition and completed six-point oral glucose tolerance tests (OGTT), Homeostatic Model Assessment of Insulin Resistance (HOMA2-IR), insulin sensitivity, detailed 3-day food records, and physical activity questionnaire. Daily habitual FODMAP intake decreased progressively with increasing BMI, ranging from 7.9 (6.2-12.7) g/d in subjects with normal BMI and 6.6 (4.6-9.9) g/d in subjects with overweight to 5.8 (3.8-9.0) g/d in subjects with obesity (p = 0.038). After adjustment for age and gender, galactooligosaccharides (GOSs) were negatively correlated with body fat (Standardised Beta coefficient β = -0.156, p = 0.006) and positively associated with insulin sensitivity (β = 0.243, p = 0.001). This remained significant after adjustment for macronutrients, fibre, and physical activity (p = 0.035 and p = 0.010, respectively). In individuals with IGT, higher dietary GOS intake was associated with lower body fat and higher insulin sensitivity independent of macronutrients and fibre intake, calling for interventional studies to evaluate the effect of FODMAP intake in prediabetes.
Collapse
Affiliation(s)
- Natural H. S. Chu
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China; (N.H.S.C.)
| | - Jie He
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China; (N.H.S.C.)
| | - Kathy H. T. Leung
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China; (N.H.S.C.)
| | - Ronald C. W. Ma
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China; (N.H.S.C.)
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Jimmy Y. S. Lee
- Department of Gastroenterology, Monash University and Alfred Hospital, Melbourne, VIC 3004, Australia
| | - Jane Varney
- Department of Gastroenterology, Monash University and Alfred Hospital, Melbourne, VIC 3004, Australia
| | - Juliana C. N. Chan
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China; (N.H.S.C.)
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Jane G. Muir
- Department of Gastroenterology, Monash University and Alfred Hospital, Melbourne, VIC 3004, Australia
| | - Elaine Chow
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China; (N.H.S.C.)
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| |
Collapse
|
36
|
Nagy DU, Sándor-Bajusz KA, Bódy B, Decsi T, Van Harsselaar J, Theis S, Lohner S. Effect of chicory-derived inulin-type fructans on abundance of Bifidobacterium and on bowel function: a systematic review with meta-analyses. Crit Rev Food Sci Nutr 2023; 63:12018-12035. [PMID: 35833477 DOI: 10.1080/10408398.2022.2098246] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Inulin-type fructans are considered to stimulate the growth of beneficial microorganisms, like Bifidobacterium in the gut and support health. However, both the fructan source and chemical structure may modify these effects. A systematic review was conducted to assess the effects of chicory-derived inulin-type fructans consumed either in specific foods or as dietary supplements on abundance of Bifidobacterium in the gut and on health-related outcomes. Three electronic databases and two clinical trial registries were systematically searched until January 2021. Two authors independently selected randomized controlled trials that investigated with a protocol of minimum seven days supplementation the effect of chicory-derived inulin-type fructans on Bifidobacterium abundance in any population. Meta-analyses with random-effects model were conducted on Bifidobacterium abundance and bowel function parameters. We evaluated risk of bias using Cochrane RoB tool. Chicory-derived inulin-type fructans at a dose of 3-20 g/day significantly increased Bifidobacterium abundance in participants with an age range from 0 to 83 years (standardized mean difference: 0.83, 95% CI: 0.58-1.08; p < 0.01; 50 studies; 2525 participants). Significant bifidogenic effects were observed in healthy individuals and in populations with health impairments, except gastrointestinal disorders. Significant beneficial effects on bowel function parameters were observed in healthy subjects. Chicory-derived inulin-type fructans may have significant bifidogenic effects and may beneficially influence bowel function in healthy individuals. PROSPERO registration number CRD42020162892.
Collapse
Affiliation(s)
- Dávid U Nagy
- Department of Paediatrics, Clinical Center of the University of Pécs, Medical School, University of Pécs, Pécs, Hungary
- Institute of Geobotany/Plant Ecology, Martin-Luther-University, Halle (Saale), Germany
| | - Kinga Amália Sándor-Bajusz
- Department of Paediatrics, Clinical Center of the University of Pécs, Medical School, University of Pécs, Pécs, Hungary
| | - Blanka Bódy
- Department of Paediatrics, Clinical Center of the University of Pécs, Medical School, University of Pécs, Pécs, Hungary
| | - Tamás Decsi
- Department of Paediatrics, Clinical Center of the University of Pécs, Medical School, University of Pécs, Pécs, Hungary
| | | | - Stephan Theis
- BENEO-Institute, c/o BENEO GmbH, Obrigheim, (Germany)
| | - Szimonetta Lohner
- Department of Public Health Medicine, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|
37
|
Basu A, Hooyman A, Richardson LA, Alman AC, Snell-Bergeon JK. Longitudinal Associations of Dietary Fiber Intake with Glycated Hemoglobin and Estimated Insulin Sensitivity in Adults with and without Type 1 Diabetes. Nutrients 2023; 15:4620. [PMID: 37960272 PMCID: PMC10648902 DOI: 10.3390/nu15214620] [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: 10/13/2023] [Revised: 10/27/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023] Open
Abstract
Dietary fiber, an essential bioactive compound in plant-based diets, is of public health concern based on habitual low intakes in the general population. Not much data are available on how habitual dietary fiber is associated with glycemic control in type 1 diabetes (T1D) as well as in prediabetes and normoglycemic adults. To address this gap, we conducted a six-year longitudinal analysis of an original cohort in adults with and without T1D (n = 1255; T1D: n = 563; non-diabetes mellitus (non-DM): n = 692). Dietary data were collected from a validated food frequency questionnaire, biochemical measures were obtained after an overnight fast, and anthropometric measurements were collected at baseline as well as after three and six years for the follow-up study. Glycated hemoglobin (HbA1c) and estimated insulin sensitivity (eIS) were the main outcomes examined. In adjusted analyses, dietary fiber intake was inversely associated with HbA1c in a minimally adjusted model, but it was positively associated with eIS in a model involving all relevant covariates in non-DM adults. These associations were not significant in the T1D group. Furthermore, when examined by HbA1c cut-offs for glycemic control, an inverse association with dietary fiber was only observed in adults with prediabetes (all p < 0.05). At a six-year mean (±SD) dietary fiber intake of 17.4 ± 8.8 g for non-DM and 17.0 ± 8.2 g for the T1D group, protective associations against poor glycemic control were observed in those without diabetes and in prediabetes.
Collapse
Affiliation(s)
- Arpita Basu
- Department of Kinesiology and Nutrition Sciences, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV 89154, USA;
| | - Andrew Hooyman
- Department of Kinesiology and Nutrition Sciences, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV 89154, USA;
- School of Biological Health Systems Engineering, Arizona State University, Tempe, AZ 85281, USA
| | - Leigh Ann Richardson
- Department of Epidemiology and Biostatistics, School of Public Health, University of Nevada Las Vegas, Las Vegas, NV 89154, USA;
| | - Amy C. Alman
- College of Public Health, University of South Florida, Tampa, FL 33620, USA;
| | - Janet K. Snell-Bergeon
- Barbara Davis Center for Diabetes, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA;
| |
Collapse
|
38
|
Williams NC, Jayaratnasingam J, Prayle AP, Nevitt SJ, Smyth AR. Prebiotics for people with cystic fibrosis. Cochrane Database Syst Rev 2023; 9:CD015236. [PMID: 37753791 PMCID: PMC10523429 DOI: 10.1002/14651858.cd015236.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
BACKGROUND Cystic fibrosis (CF) is a multisystem disease; the importance of growth and nutritional status is well established given their implications for lung function and overall survivability. Furthermore, it has been established that intestinal microbial imbalance and inflammation are present in people with CF. Oral prebiotics are commercially available substrates that are selectively utilised by host intestinal micro-organisms and may improve both intestinal and overall health. OBJECTIVES To evaluate the benefits and harms of prebiotics for improving health outcomes in children and adults with CF. SEARCH METHODS We searched the Cochrane Cystic Fibrosis Trials Register compiled from electronic database searches and handsearching of journals and conference abstract books. We also searched the reference lists of relevant articles and reviews. Date of last search: 19 October 2022. We also searched PubMed and online trials registries. Date of last search: 13 January 2023. SELECTION CRITERIA Randomised controlled trials (RCTs) and quasi-RCTs assessing the efficacy of prebiotics in children and adults with CF. We planned to only include the first treatment period from cross-over RCTs, regardless of washout period. DATA COLLECTION AND ANALYSIS We did not identify any relevant trials. MAIN RESULTS We did not identify any relevant trials for inclusion in this review. AUTHORS' CONCLUSIONS This review did not find any evidence for the use of prebiotics in people with CF. Until such evidence is available, it is reasonable for clinicians to follow any local guidelines and to discuss the use of dietary prebiotics with their patients. Large and robust RCTs assessing the dietary prebiotics of inulin or galacto-oligosaccharides or fructo-oligosaccharides, or any combination of these, are needed. Such studies should be of at least 12 months in duration and assess outcomes such as growth and nutrition, gastrointestinal symptoms, pulmonary exacerbations, lung function, inflammatory biomarkers, hospitalisations, intestinal microbial profiling, and faecal short-chain fatty acids. Trials should include both children and adults and aim to be adequately powered to allow for subgroup analysis by age.
Collapse
Affiliation(s)
- Neil C Williams
- Exercise and Health Research Group, Sport Health and Performance Enhancement (SHAPE) Research Centre, School of Science and Technology., Nottingham Trent University, Nottingham, UK
| | - Jacob Jayaratnasingam
- Exercise and Health Research Group, Sport Health and Performance Enhancement (SHAPE) Research Centre, School of Science and Technology., Nottingham Trent University, Nottingham, UK
| | - Andrew P Prayle
- Nottingham Cystic Fibrosis and Chidlren's Respiratory Research Centre, University of Nottingham, Nottingham, UK
| | - Sarah J Nevitt
- Department of Health Data Science, University of Liverpool, Liverpool, UK
| | - Alan R Smyth
- Division of Child Health, Obstetrics & Gynaecology (COG), School of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
39
|
Kilburn-Kappeler LR, Doerksen T, Lu A, Palinski RM, Lu N, Aldrich CG. Comparison of the Effect of Corn-fermented Protein and Traditional Ingredients on the Fecal Microbiota of Dogs. Vet Sci 2023; 10:553. [PMID: 37756074 PMCID: PMC10536651 DOI: 10.3390/vetsci10090553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/28/2023] Open
Abstract
Corn-fermented protein (CFP), a co-product from the ethanol industry, is produced using post-fermentation technology to split the protein and yeast from fiber prior to drying. The objective of this study was to determine the effect of CFP compared to traditional ingredients on the fecal microbiota of dogs. The four experimental diets included a control with no yeast and diets containing either 3.5% brewer's dried yeast, 2.5% brewer's dried yeast plus 17.5% distiller's dried grains with solubles, or 17.5% CFP. The experimental diets were fed to adult dogs (n = 12) in a 4 × 4 replicated Latin square design. Fresh fecal samples (n = 48) were analyzed by 16S metagenomic sequencing. Raw sequences were processed through mothur. Community diversity was evaluated in R. Relative abundance data were analyzed within the 50 most abundant operational taxonomic units using a mixed model of SAS. Alpha and beta diversity were similar for all treatments. Predominant phyla among all samples were Firmicutes (73%), Bacteroidetes (15%), Fusobacteria (8%), and Actinobacteria (4%). There were no quantifiable (p > 0.05) shifts in the predominant phyla among the treatments. However, nine genera resulted in differences in relative abundance among the treatments. These data indicate that compared to traditional ingredients, CFP did not alter the overall diversity of the fecal microbiota of healthy adult dogs over 14 days.
Collapse
Affiliation(s)
| | - Tyler Doerksen
- Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (T.D.); (A.L.); (R.M.P.)
| | - Andrea Lu
- Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (T.D.); (A.L.); (R.M.P.)
| | - Rachel M. Palinski
- Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (T.D.); (A.L.); (R.M.P.)
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Nanyan Lu
- Bioinformatics Center, Kansas State University, Manhattan, KS 66506, USA;
| | - Charles G. Aldrich
- Department of Grain Science and Industry, Kansas State University, Manhattan, KS 66506, USA;
| |
Collapse
|
40
|
Sun F, Yang CL, Wang FX, Rong SJ, Luo JH, Lu WY, Yue TT, Wang CY, Liu SW. Pancreatic draining lymph nodes (PLNs) serve as a pathogenic hub contributing to the development of type 1 diabetes. Cell Biosci 2023; 13:156. [PMID: 37641145 PMCID: PMC10464122 DOI: 10.1186/s13578-023-01110-7] [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: 03/08/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023] Open
Abstract
Type 1 diabetes (T1D) is a chronic, progressive autoinflammatory disorder resulting from the breakdown of self-tolerance and unrestrained β cell-reactive immune response. Activation of immune cells is initiated in islet and amplified in lymphoid tissues, especially those pancreatic draining lymph nodes (PLNs). The knowledge of PLNs as the hub of aberrant immune response is continuously being replenished and renewed. Here we provide a PLN-centered view of T1D pathogenesis and emphasize that PLNs integrate signal inputs from the pancreas, gut, viral infection or peripheral circulation, undergo immune remodeling within the local microenvironment and export effector cell components into pancreas to affect T1D progression. In accordance, we suggest that T1D intervention can be implemented by three major ways: cutting off the signal inputs into PLNs (reduce inflammatory β cell damage, enhance gut integrity and control pathogenic viral infections), modulating the immune activation status of PLNs and blocking the outputs of PLNs towards pancreatic islets. Given the dynamic and complex nature of T1D etiology, the corresponding intervention strategy is thus required to be comprehensive to ensure optimal therapeutic efficacy.
Collapse
Affiliation(s)
- Fei Sun
- Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chun-Liang Yang
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fa-Xi Wang
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan-Jie Rong
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia-Hui Luo
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wan-Ying Lu
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tian-Tian Yue
- Devision of Nutrition, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cong-Yi Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China.
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Shi-Wei Liu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
41
|
Bester A, O'Brien M, Cotter PD, Dam S, Civai C. Shotgun Metagenomic Sequencing Revealed the Prebiotic Potential of a Fruit Juice Drink with Fermentable Fibres in Healthy Humans. Foods 2023; 12:2480. [PMID: 37444219 DOI: 10.3390/foods12132480] [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: 04/10/2023] [Revised: 05/13/2023] [Accepted: 06/08/2023] [Indexed: 07/15/2023] Open
Abstract
Fibre-based dietary interventions are at the forefront of gut microbiome modulation research, with a wealth of 16S rRNA information to demonstrate the prebiotic effects of isolated fibres. However, there is a distinct lack of data relating to the effect of a combination of soluble and insoluble fibres in a convenient-to-consume fruit juice food matrix on gut microbiota structure, diversity, and function. Here, we aimed to determine the impact of the MOJU Prebiotic Shot, an apple, lemon, ginger, and raspberry fruit juice drink blend containing chicory inulin, baobab, golden kiwi, and green banana powders, on gut microbiota structure and function. Healthy adults (n = 20) were included in a randomised, double-blind, placebo-controlled, cross-over study, receiving 60 mL MOJU Prebiotic Shot or placebo (without the fibre mix) for 3 weeks with a 3-week washout period between interventions. Shotgun metagenomics revealed significant between-group differences in alpha and beta diversity. In addition, the relative abundance of the phyla Actinobacteria and Desulfobacteria was significantly increased as a result of the prebiotic intervention. Nine species were observed to be differentially abundant (uncorrected p-value of <0.05) as a result of the prebiotic treatment. Of these, Bifidobacterium adolescentis and CAG-81 sp900066785 (Lachnospiraceae) were present at increased abundance relative to baseline. Additionally, KEGG analysis showed an increased abundance in pathways associated with arginine biosynthesis and phenylacetate degradation during the prebiotic treatment. Our results show the effects of the daily consumption of 60 mL MOJU Prebiotic Shot for 3 weeks and provide insight into the functional potential of B. adolescentis.
Collapse
Affiliation(s)
- Adri Bester
- London Agri Food Innovation Clinic (LAFIC), School of Applied Sciences, London South Bank University, London SE1 0AA, UK
| | | | | | | | - Claudia Civai
- London Agri Food Innovation Clinic (LAFIC), School of Applied Sciences, London South Bank University, London SE1 0AA, UK
| |
Collapse
|
42
|
Ogrotis I, Koufakis T, Kotsa K. Changes in the Global Epidemiology of Type 1 Diabetes in an Evolving Landscape of Environmental Factors: Causes, Challenges, and Opportunities. Medicina (B Aires) 2023; 59:medicina59040668. [PMID: 37109626 PMCID: PMC10141720 DOI: 10.3390/medicina59040668] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/25/2023] [Accepted: 03/27/2023] [Indexed: 03/30/2023] Open
Abstract
The worldwide incidence of type 1 diabetes mellitus (T1DM) has increased in recent decades. The reasons behind this phenomenon are not yet fully understood. Early life infections, prenatal and perinatal factors, and diet composition have been associated with the triggering of autoimmunity and the risk of presentation of T1DM. However, the rapid increase in new cases of the disease raises the hypothesis that lifestyle factors, which have traditionally been associated with type 2 diabetes, such as obesity and unhealthy eating patterns could also play a role in the genesis of autoimmune diabetes. This article aims to highlight the changing epidemiology of T1DM and the importance of properly recognizing the environmental factors behind it, as well as the connections with the pathogenesis of the disorder and the need to prevent or delay T1DM and its long-term complications.
Collapse
Affiliation(s)
- Ioannis Ogrotis
- School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Theocharis Koufakis
- Division of Endocrinology and Metabolism and Diabetes Center, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, 54636 Thessaloniki, Greece
| | - Kalliopi Kotsa
- Division of Endocrinology and Metabolism and Diabetes Center, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, 54636 Thessaloniki, Greece
- Correspondence: ; Tel.: +30-231-099-4706
| |
Collapse
|
43
|
Piccioni A, Covino M, Candelli M, Ojetti V, Capacci A, Gasbarrini A, Franceschi F, Merra G. How Do Diet Patterns, Single Foods, Prebiotics and Probiotics Impact Gut Microbiota? MICROBIOLOGY RESEARCH 2023; 14:390-408. [DOI: 10.3390/microbiolres14010030] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2023] Open
Abstract
The human gastrointestinal tract hosts a complex and dynamic population of commensal bacterial species, which have coevolved with the host, generating a symbiotic relationship. Some compounds present in foods, such as polyols, prebiotic fibers, or phenolic compounds, are poorly metabolized and absorbed by the host before the transformation guided by the colonic microbiota. By influencing gut microbiota, diet plays a fundamental role in understanding the beneficial effects of the gut microbiota on the host, including its long-term metabolism. The idea that probiotics can act not only by influencing the colonizing microbiota opens the door to a wider range of probiotic possibilities, encouraging innovation in the field. Furthermore, it has been shown both that some probiotics increase phagocytosis or the activity of natural killer cells. Current prebiotics are mainly based on carbohydrates, but other substances, such as polyphenols and polyunsaturated fatty acids, could exert prebiotic effects. A prebiotic substance has been defined as ‘a substrate that is selectively used by host microorganisms that confer a health benefit’, and so can interact with the gut microbiota through competition for nutrients, antagonism, cross-feeding, and support for microbiota stability. Influencing its composition in terms of richness and diversity, food components have a key impact on the intestinal microbiota. Eating habits can strongly influence the composition of the intestinal microbiota. A healthy intestinal microbiota is essential for maintaining general health, and diet is one of the major modulators of this fascinating world of microorganisms. This must give us one more reason to adopt a healthy lifestyle.
Collapse
Affiliation(s)
- Andrea Piccioni
- Department of Emergency Medicine, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Marcello Covino
- Department of Emergency Medicine, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Marcello Candelli
- Department of Emergency Medicine, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Veronica Ojetti
- Department of Emergency Medicine, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Annunziata Capacci
- Department of Medical and Surgical Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Department of Medical and Surgical Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Francesco Franceschi
- Department of Emergency Medicine, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Giuseppe Merra
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00136 Rome, Italy
| |
Collapse
|
44
|
Rubin D, Bosy-Westphal A, Kabisch S, Kronsbein P, Simon MC, Tombek A, Weber KS, Skurk T. Nutritional Recommendations for People with Type 1 Diabetes Mellitus. Exp Clin Endocrinol Diabetes 2023; 131:33-50. [PMID: 36638807 DOI: 10.1055/a-1946-3753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Diana Rubin
- Vivantes Hospital Spandau, Berlin, Germany.,Vivantes Humboldt Hospital, Berlin, Germany
| | - Anja Bosy-Westphal
- Institute of Human Nutrition, Faculty of Agriculture and Nutritional Sciences, Christian-Albrechts University of Kiel, Kiel, Germany
| | - Stefan Kabisch
- Department of Endocrinology, Diabetes and Nutritional Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany.,German Center for Diabetes Research (DZD), Munich, Germany
| | - Peter Kronsbein
- Faculty of Nutrition and Food Sciences, Niederrhein University of Applied Sciences, Mönchengladbach, Germany
| | - Marie-Christine Simon
- Institute of Nutrition and Food Sciences, Rhenish Friedrich Wilhelm University of Bonn, Bonn, Germany
| | - Astrid Tombek
- Diabetes Center Bad Mergentheim, Bad Mergentheim, Germany
| | - Katharina S Weber
- Institute for Epidemiology, Christian-Albrechts University of Kiel, Kiel, Germany
| | - Thomas Skurk
- ZIEL - Institute for Food & Health, Technical University Munich, Freising, Germany
| |
Collapse
|
45
|
Liu B, Zhang L, Yang H, Zheng H, Liao X. Microbiota: A potential orchestrator of antidiabetic therapy. Front Endocrinol (Lausanne) 2023; 14:973624. [PMID: 36777348 PMCID: PMC9911464 DOI: 10.3389/fendo.2023.973624] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 01/17/2023] [Indexed: 01/28/2023] Open
Abstract
The gut microbiota, as a 'new organ' of humans, has been identified to affect many biological processes, including immunity, inflammatory response, gut-brain neural circuits, and energy metabolism. Profound dysbiosis of the gut microbiome could change the metabolic pattern, aggravate systemic inflammation and insulin resistance, and exacerbate metabolic disturbance and the progression of type 2 diabetes (T2D). The aim of this review is to focus on the potential roles and functional mechanisms of gut microbiota in the antidiabetic therapy. In general, antidiabetic drugs (α-glucosidase inhibitor, biguanides, incretin-based agents, and traditional Chinese medicine) induce the alteration of microbial diversity and composition, and the levels of bacterial component and derived metabolites, such as lipopolysaccharide (LPS), short chain fatty acids (SCFAs), bile acids and indoles. The altered microbial metabolites are involved in the regulation of gut barrier, inflammation response, insulin resistance and glucose homeostasis. Furthermore, we summarize the new strategies for antidiabetic treatment based on microbial regulation, such as pro/prebiotics administration and fecal microbiota transplantation, and discuss the need for more basic and clinical researches to evaluate the feasibility and efficacy of the new therapies for diabetes.
Collapse
Affiliation(s)
| | | | | | - Hongting Zheng
- Department of Endocrinology, Chongqing Education Commission Key Laboratory of Diabetic Translational Research, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Xiaoyu Liao
- Department of Endocrinology, Chongqing Education Commission Key Laboratory of Diabetic Translational Research, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| |
Collapse
|
46
|
Campbell C, Kandalgaonkar MR, Golonka RM, Yeoh BS, Vijay-Kumar M, Saha P. Crosstalk between Gut Microbiota and Host Immunity: Impact on Inflammation and Immunotherapy. Biomedicines 2023; 11:294. [PMID: 36830830 PMCID: PMC9953403 DOI: 10.3390/biomedicines11020294] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/09/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
Gut microbes and their metabolites are actively involved in the development and regulation of host immunity, which can influence disease susceptibility. Herein, we review the most recent research advancements in the gut microbiota-immune axis. We discuss in detail how the gut microbiota is a tipping point for neonatal immune development as indicated by newly uncovered phenomenon, such as maternal imprinting, in utero intestinal metabolome, and weaning reaction. We describe how the gut microbiota shapes both innate and adaptive immunity with emphasis on the metabolites short-chain fatty acids and secondary bile acids. We also comprehensively delineate how disruption in the microbiota-immune axis results in immune-mediated diseases, such as gastrointestinal infections, inflammatory bowel diseases, cardiometabolic disorders (e.g., cardiovascular diseases, diabetes, and hypertension), autoimmunity (e.g., rheumatoid arthritis), hypersensitivity (e.g., asthma and allergies), psychological disorders (e.g., anxiety), and cancer (e.g., colorectal and hepatic). We further encompass the role of fecal microbiota transplantation, probiotics, prebiotics, and dietary polyphenols in reshaping the gut microbiota and their therapeutic potential. Continuing, we examine how the gut microbiota modulates immune therapies, including immune checkpoint inhibitors, JAK inhibitors, and anti-TNF therapies. We lastly mention the current challenges in metagenomics, germ-free models, and microbiota recapitulation to a achieve fundamental understanding for how gut microbiota regulates immunity. Altogether, this review proposes improving immunotherapy efficacy from the perspective of microbiome-targeted interventions.
Collapse
Affiliation(s)
- Connor Campbell
- Department of Physiology & Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Mrunmayee R. Kandalgaonkar
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Rachel M. Golonka
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Beng San Yeoh
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Matam Vijay-Kumar
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Piu Saha
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| |
Collapse
|
47
|
Vazquez-Marroquin G, Ochoa-Précoma R, Porchia LM, Pérez-Fuentes R, Nicolás-Toledo L, Rodríguez-Antolín J, Gonzalez-Mejia ME. The Effect of Microbiome Therapies on Waist Circumference, a Measure of Central Obesity, in Patients with Type 2 Diabetes: A Systematic Review and Meta-analysis of Randomized Controlled Trials. J Acad Nutr Diet 2023; 123:933-952.e1. [PMID: 36634870 DOI: 10.1016/j.jand.2023.01.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/11/2023]
Abstract
BACKGROUND Microbiome therapies (probiotic, prebiotic, and synbiotics) have been proposed as adjuvants in the control of central obesity; however, their results for patients with type 2 diabetes (T2D) remain inconclusive. OBJECTIVE The aim of this systematic review and meta-analysis was to evaluate the effect of microbiome therapies on central obesity as measured by waist circumference (WC), and to evaluate the effect of microbiome therapies for glycemic parameters (fasting glucose [FPG], fasting insulin [FPI], hemoglobin A1c [HbA1c], and insulin resistance [HOMA1-IR]) in patients with T2D. METHODS SCOPUS, Pubmed, EBSCO, and LILACS databases were searched for studies that investigated the effect of microbiome therapies on WC up to June 1, 2022. Heterogeneity was determined using Cochran's Q test and quantified using the inconsistency index. The random effects model was used to calculate the pooled difference in means (DM) and 95% confidence intervals (95%CI). Egger's test and Beggs-Muzamar's test were used to assess publication bias. RESULTS Fifteen reports were included (443 treated and 387 controls). Overall, a significant decrease in WC was found (DM = -0.97 cm; 95% confidence interval [95%CI] = -1.74 to -0.20; P = 0.014); however, when stratified by type of microbiome therapy, only probiotics significantly decreased WC (DM = -0.62 cm; 95%CI = -1.00 to -0.24; P = 0.002). No effect was observed for prebiotics and synbiotics. With respect to glycemic parameters, HbA1c, FPG, and HOMA1-IR significantly decrease with microbiome therapies (P ≤ 0.001). When stratified by the type of therapy, for probiotic treatments, HbA1c, FPG, and HOMA1-IR scores decrease (P < 0.001). For prebiotic treatments, HbA1c and FPG (P ≤ 0.001) levels decrease, whereas FPI increased (P = 0.012). Synbiotic treatments were only associated with an increase in FPI (P = 0.031). CONCLUSION Findings indicate that using probiotics alone improved WC in patients with T2D. Both probiotics and prebiotics decreased HbA1c and FPG; however, prebiotics and synbiotics resulted in an increase in FPI. The formulation of the therapy (single vs multi) had no difference on the effect.
Collapse
|
48
|
Peñalver Bernabé B, Maki PM, Cunningham JL, Eisenlohr-Moul T, Tussing-Humphreys L, Carroll IM, Meltzer-Brody S, Gilbert JA, Kimmel M. Interactions between perceived stress and microbial-host immune components: two demographically and geographically distinct pregnancy cohorts. Transl Psychiatry 2023; 13:3. [PMID: 36609477 PMCID: PMC9822983 DOI: 10.1038/s41398-022-02276-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 11/18/2022] [Accepted: 11/30/2022] [Indexed: 01/09/2023] Open
Abstract
Higher stress during pregnancy associates with negative outcomes and elevated inflammation. The gut microbiota, reflecting environment and social interactions, alongside host immune responses have the potential to better understand perceived stress and identify when stress is excessive in pregnancy. Two U.S. cohorts of 84 pregnant individuals, composed of urban women of color and suburban white women, completed the Perceived Stress Scale-10 (PSS-10) and provided fecal and blood samples at two time points. Confirmatory Factor Analysis assessed the robustness of a two-factor PSS-10 model (Emotional Distress/ED and Self-Efficacy/SE). Gut microbiota composition was measured by 16 S rRNA amplicon sequencing and the immune system activity was assessed with a panel of 21 T-cell related cytokines and chemokines. ED levels were higher in the suburban compared to the urban cohort, but levels of SE were similar. ED and SE levels were associated with distinct taxonomical signatures and the gut microbiota data improved the prediction of SE levels compared with models based on socio-demographic characteristics alone. Integration of self-reported symptoms, microbial and immune information revealed a possible mediation effect of Bacteroides uniformis between the immune system (through CXCL11) and SE. The study identified links between distinct taxonomical and immunological signatures with perceived stress. The data are congruent with a model where gut microbiome and immune factors, both impacting and reflecting factors such as close social relationships and dietary fiber, may modulate neural plasticity resulting in increased SE during pregnancy. The predictive value of these peripheral markers merit further study.
Collapse
Affiliation(s)
- Beatriz Peñalver Bernabé
- Department of Biomedical Engineering, College of Engineering and Medicine, University of Illinois Chicago, Chicago, IL, USA. .,Center of Bioinformatics and Quantitative Biology, University of Illinois Chicago, Chicago, IL, USA.
| | - Pauline M. Maki
- grid.185648.60000 0001 2175 0319Department of Psychology, College of Medicine, University of Illinois Chicago, Chicago, IL USA ,grid.185648.60000 0001 2175 0319Department of Psychiatry, College of Medicine, University of Illinois Chicago, Chicago, IL USA ,grid.185648.60000 0001 2175 0319Department of Obstetrics and Gynecology, College of Medicine, University of Illinois Chicago, Chicago, IL USA
| | - Janet L. Cunningham
- grid.8993.b0000 0004 1936 9457Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden
| | - Tory Eisenlohr-Moul
- grid.185648.60000 0001 2175 0319Department of Psychology, College of Medicine, University of Illinois Chicago, Chicago, IL USA ,grid.185648.60000 0001 2175 0319Department of Psychiatry, College of Medicine, University of Illinois Chicago, Chicago, IL USA
| | - Lisa Tussing-Humphreys
- grid.185648.60000 0001 2175 0319Department of Kinesiology and Nutrition, College of Applied Health Sciences, University of Illinois Chicago, Chicago, IL USA
| | - Ian M. Carroll
- grid.410711.20000 0001 1034 1720Department of Nutrition, School of Public Health, University of North Carolina, Chapel Hill, NC USA
| | - Samantha Meltzer-Brody
- grid.410711.20000 0001 1034 1720Department of Psychiatry, University of North Carolina, Chapel Hill, NC USA
| | - Jack A. Gilbert
- grid.266100.30000 0001 2107 4242Department of Pediatrics and Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA USA
| | - Mary Kimmel
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA. .,Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
49
|
Shen R, Chen S, Lei W, Shen J, Lv L, Wei T. Nonfood Probiotic, Prebiotic, and Synbiotic Use Reduces All-Cause and Cardiovascular Mortality Risk in Older Adults: A Population-Based Cohort Study. J Nutr Health Aging 2023; 27:391-397. [PMID: 37248763 DOI: 10.1007/s12603-023-1921-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 04/14/2023] [Indexed: 05/31/2023]
Abstract
OBJECTIVES Pro-, pre-, and synbiotic supplements improve cardiovascular risk factors. However, the association between nonfood pro-, pre-, and synbiotics (NPPS) and long-term all-cause and cardiovascular mortality has not been studied. Thus, our objective was to determine the impact of nonfood pro-, pre-, and synbiotics on all-cause and cardiovascular mortality. DESIGN, SETTING, AND PARTICIPANTS This was a retrospective, cohort study of 4837 nationally representative American participants aged 65 years or older with a median follow-up duration of 77 months. MEASUREMENTS All-cause and cardiovascular mortality were measured. RESULTS A total of 1556 participants died during the median 77-month follow-up, and 517 died from cardiovascular disease. Compared with participants without NPPS use, participants who used NPPS experienced a reduced risk of all-cause mortality by nearly 41% (hazard ratio 0.59, 95% CI 0.43 to 0.79) and cardiovascular mortality by 52% (HR 0.48, 95% CI 0.30 to 0.76). Such an effect persisted in most subgroup analyses and complete-case analyses. CONCLUSION AND RELEVANCE In this study, we found a protective effect of NPPS against all-cause and cardiovascular mortality in Americans aged 65 years or older. Nonfood pro-, pre-, and synbiotics can be a novel, inexpensive, low-risk treatment addition for all-cause and cardiovascular mortality for older individuals.
Collapse
Affiliation(s)
- R Shen
- Tiemin Wei, Department of Cardiology, Lishui Hospital, Zhejiang University School of Medicine, No.289, Kuocang Road, Liandu District, Lishui, China. Tel: 86+139 0588 7981, . Co-corresponding author: Lingchun Lv, E-mail:
| | | | | | | | | | | |
Collapse
|
50
|
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
|