1
|
Zhang S, Ma J, Ma Y, Yi J, Wang B, Wang H, Yang Q, Zhang K, Yan X, Sun D, You J. Engineering Probiotics for Diabetes Management: Advances, Challenges, and Future Directions in Translational Microbiology. Int J Nanomedicine 2024; 19:10917-10940. [PMID: 39493275 PMCID: PMC11530765 DOI: 10.2147/ijn.s492651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 10/08/2024] [Indexed: 11/05/2024] Open
Abstract
Background Diabetes Mellitus (DM) is a substantial health concern worldwide, and its incidence is progressively escalating. Conventional pharmacological interventions frequently entail undesirable side effects, and while probiotics offer benefits, they are hindered by constraints such as diminished stability and effectiveness within the gastrointestinal milieu. Given these complications, the advent of bioengineered probiotics is a promising alternative for DM management. Aim of Review The objective of this review is to provide an exhaustive synthesis of the most recent studies on the use of engineered probiotics in the management of DM. This study aimed to clarify the mechanisms through which these probiotics function, evaluate their clinical effectiveness, and enhance public awareness of their prospective advantages in the treatment of DM. Key Scientific Concepts of Review Scholarly critiques have explored diverse methodologies of probiotic engineering, including physical alteration, bioenrichment, and genetic manipulation. These techniques augment the therapeutic potency of probiotics by ameliorating gut microbiota, fortifying the intestinal barrier, modulating metabolic pathways, and regulating immune responses. Such advancements have established engineered probiotics as a credible therapeutic strategy for DM, potentially providing enhanced results compared to conventional treatments.
Collapse
Affiliation(s)
- Shenghao Zhang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, People’s Republic of China
| | - Jiahui Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, People’s Republic of China
| | - Yilei Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, People’s Republic of China
| | - Jia Yi
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, People’s Republic of China
| | - Beier Wang
- Department of Hepatobiliary-Pancreatic Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People’s Republic of China
| | - Hanbing Wang
- Department of Biotechnology, The University of Hong Kong, Hong Kong SAR, 999077, People’s Republic of China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, People’s Republic of China
| | - Kun Zhang
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, 404000, People’s Republic of China
| | - Xiaoqing Yan
- The Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, People’s Republic of China
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, People’s Republic of China
- Department of Endocrinology, Yiwu Central Hospital, The Affiliated Yiwu hospital of Wenzhou Medical University, Yiwu, 322000, People’s Republic of China
| | - Jinfeng You
- Department of Obstetrics, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, 324000, People’s Republic of China
| |
Collapse
|
2
|
Tegegne BA, Adugna A, Yenet A, Yihunie Belay W, Yibeltal Y, Dagne A, Hibstu Teffera Z, Amare GA, Abebaw D, Tewabe H, Abebe RB, Zeleke TK. A critical review on diabetes mellitus type 1 and type 2 management approaches: from lifestyle modification to current and novel targets and therapeutic agents. Front Endocrinol (Lausanne) 2024; 15:1440456. [PMID: 39493778 PMCID: PMC11527681 DOI: 10.3389/fendo.2024.1440456] [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: 05/29/2024] [Accepted: 10/02/2024] [Indexed: 11/05/2024] Open
Abstract
Diabetes mellitus (DM) has emerged as an international health epidemic due to its rapid rise in prevalence. Consequently, scientists and or researchers will continue to find novel, safe, effective, and affordable anti-diabetic medications. The goal of this review is to provide a thorough overview of the role that lifestyle changes play in managing diabetes, as well as the standard medications that are currently being used to treat the condition and the most recent advancements in the development of novel medical treatments that may be used as future interventions for the disease. A literature search was conducted using research databases such as PubMed, Web of Science, Scopus, ScienceDirect, Wiley Online Library, Google Scholar, etc. Data were then abstracted from these publications using words or Phrases like "pathophysiology of diabetes", "Signe and symptoms of diabetes", "types of diabetes", "major risk factors and complication of diabetes", "diagnosis of diabetes", "lifestyle modification for diabetes", "current antidiabetic agents", and "novel drugs and targets for diabetes management" that were published in English and had a strong scientific foundation. Special emphasis was given to the importance of lifestyle modification, as well as current, novel, and emerging/promising drugs and targets helpful for the management of both T1DM and T2DM.
Collapse
Affiliation(s)
- Bantayehu Addis Tegegne
- Department of Pharmacy, College of Medicine and Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Adane Adugna
- Department of Medical Laboratory Science, College of Medicine and Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Aderaw Yenet
- Department of Pharmacy, College of Medicine and Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Wubetu Yihunie Belay
- Department of Pharmacy, College of Medicine and Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Yared Yibeltal
- Department of Pharmacy, College of Medicine and Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Abebe Dagne
- Department of Pharmacy, College of Medicine and Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Zigale Hibstu Teffera
- Department of Medical Laboratory Science, College of Medicine and Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Gashaw Azanaw Amare
- Department of Medical Laboratory Science, College of Medicine and Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Desalegn Abebaw
- Department of Medical Laboratory Science, College of Medicine and Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Haymanot Tewabe
- Department of Medical Laboratory Science, College of Medicine and Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Rahel Belete Abebe
- Department of Clinical Pharmacy, School of Pharmacy, College of Medicine and Health Science, University of Gondar, Gondar, Ethiopia
| | - Tirsit Ketsela Zeleke
- Department of Pharmacy, College of Medicine and Health Science, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
3
|
Prapa I, Kompoura V, Pavlatou C, Nelios G, Mitropoulou G, Kostomitsopoulos N, Plessas S, Bezirtzoglou E, Karathanos VT, Yanni AE, Kourkoutas Y. Effects of Free or Immobilized Pediococcus acidilactici ORE5 on Corinthian Currants on Gut Microbiome of Streptozotocin-Induced Diabetic Rats. Microorganisms 2024; 12:2004. [PMID: 39458313 PMCID: PMC11509866 DOI: 10.3390/microorganisms12102004] [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: 08/08/2024] [Revised: 09/29/2024] [Accepted: 09/29/2024] [Indexed: 10/28/2024] Open
Abstract
The present study aimed to investigate the effect of a dietary intervention including free or immobilized cells of the presumptive probiotic Pediococcus acidilactici ORE5 on Corinthian currants, a food with beneficial impact in the condition of Type-1 Diabetes Mellitus (T1DM), on the microbiome composition of STZ-induced diabetic rats. Twenty four male Wistar rats were divided into four groups (n = 6 per group): healthy animals, which received the free (H_FP) or the immobilized Pediococcus acidilactici ORE5 cells (H_IPC), and diabetic animals, which received the free (D_FP) or the immobilized Pediococcus acidilactici ORE5 cells(D_IPC) for 4 weeks (109 cfu/day, in all groups). At the end of the dietary intervention, the D_IPC group exerted a lower concentration of the inflammatory cytokine IL-1 beta compared to D_FP. Consumption of immobilized P. acidilactici ORE5 cells on Corinthian currants by diabetic animals led to increased loads of fecal lactobacilli and lower Enterobacteriaceae, coliforms, and Escherichia coli levels, while Actinobacteria phylum, Akkermansia, and Bifidobacterium genera abundances were increased, and fecal lactic acid was elevated. Overall, the results of the present research demonstrated that functional ingredients could ameliorate gut dysbiosis present in T1DM and could be used to design dietary patterns aiming at T1DM management. However, well-designed clinical trials are necessary, in order to confirm the beneficial effects in humans.
Collapse
Affiliation(s)
- Ioanna Prapa
- Laboratory of Applied Microbiology and Biotechnology, Department of Molecular Biology and Genetics, Democritus University of Thrace, Dragana, 68100 Alexandroupolis, Greece; (I.P.); (V.K.); (C.P.); (G.N.); (G.M.)
| | - Vasiliki Kompoura
- Laboratory of Applied Microbiology and Biotechnology, Department of Molecular Biology and Genetics, Democritus University of Thrace, Dragana, 68100 Alexandroupolis, Greece; (I.P.); (V.K.); (C.P.); (G.N.); (G.M.)
| | - Chrysoula Pavlatou
- Laboratory of Applied Microbiology and Biotechnology, Department of Molecular Biology and Genetics, Democritus University of Thrace, Dragana, 68100 Alexandroupolis, Greece; (I.P.); (V.K.); (C.P.); (G.N.); (G.M.)
| | - Grigorios Nelios
- Laboratory of Applied Microbiology and Biotechnology, Department of Molecular Biology and Genetics, Democritus University of Thrace, Dragana, 68100 Alexandroupolis, Greece; (I.P.); (V.K.); (C.P.); (G.N.); (G.M.)
| | - Gregoria Mitropoulou
- Laboratory of Applied Microbiology and Biotechnology, Department of Molecular Biology and Genetics, Democritus University of Thrace, Dragana, 68100 Alexandroupolis, Greece; (I.P.); (V.K.); (C.P.); (G.N.); (G.M.)
| | - Nikolaos Kostomitsopoulos
- Laboratory Animal Facility, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece;
| | - Stavros Plessas
- Laboratory of Microbiology, Biotechnology and Hygiene, Faculty of Agricultural Development, Democritus University of Thrace, 68200 Orestiada, Greece;
| | - Eugenia Bezirtzoglou
- Laboratory of Hygiene and Environmental Protection, Department of Medicine, Democritus University of Thrace, Dragana, 68100 Alexandroupolis, Greece;
| | - Vaios T. Karathanos
- Laboratory of Chemistry, Biochemistry, Physical Chemistry of Foods, Department of Nutrition and Dietetics, Harokopio University of Athens, 17671 Athens, Greece;
- Agricultural Cooperatives’ Union of Aeghion, Corinthou 201, 25100 Aeghion, Greece
| | - Amalia E. Yanni
- Laboratory of Chemistry, Biochemistry, Physical Chemistry of Foods, Department of Nutrition and Dietetics, Harokopio University of Athens, 17671 Athens, Greece;
| | - Yiannis Kourkoutas
- Laboratory of Applied Microbiology and Biotechnology, Department of Molecular Biology and Genetics, Democritus University of Thrace, Dragana, 68100 Alexandroupolis, Greece; (I.P.); (V.K.); (C.P.); (G.N.); (G.M.)
| |
Collapse
|
4
|
Bajinka O, Sylvain Dovi K, Simbilyabo L, Conteh I, Tan Y. The predicted mechanisms and evidence of probiotics on type 2 diabetes mellitus (T2DM). Arch Physiol Biochem 2024; 130:475-490. [PMID: 36630122 DOI: 10.1080/13813455.2022.2163260] [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: 09/10/2022] [Revised: 12/01/2022] [Accepted: 12/15/2022] [Indexed: 01/12/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is a serious endocrine and metabolic disease that is highly prevalent and causes high mortality and morbidity rates worldwide. This review aims to focus on the potential of probiotics in the management of T2DM and its complications and to summarise the various mechanisms of action of probiotics with respect to T2DM. In this review, experimental studies conducted between 2016 and 2022 were explored. The possible mechanisms of action are based on their ability to modulate the gut microbiota, boost the production of short-chain fatty acids (SCFAs) and glucagon-like peptides, inhibit α-glucosidase, elevate sirtuin 1 (SIRT1) levels while reducing fetuin-A levels, and regulate the level of inflammatory cytokines. This review recommends carrying out further studies, especially human trials, to provide robust evidence-based knowledge on the use of probiotics for the treatment of T2DM.IMPACT STATEMENTT2DM is prevalent worldwide causing high rates of morbidity and mortality.Gut microbiota play a significant role in the pathogenesis of T2DM.Probiotics can be used as possible therapeutic tools for the management of T2DM.The possible mechanisms of action of probiotics include modulation of the gut microbiota, production of SCFAs and glucagon-like peptides, inhibition of α-glucosidase, raising SIRT1, reducing fetuin-A levels, and regulating the level of inflammatory cytokines.
Collapse
Affiliation(s)
- Ousman Bajinka
- Department of Medical Microbiology, Central South University, Changsha, China
- China-Africa Research Center of Infectious Diseases, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Kodzovi Sylvain Dovi
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, P. R. China
| | - Lucette Simbilyabo
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, China
| | - Ishmail Conteh
- Department of Epidemiology and Health Statistics, Xiangya School of public health central South University, Changsha, P. R. China
| | - Yurong Tan
- Department of Medical Microbiology, Central South University, Changsha, China
- China-Africa Research Center of Infectious Diseases, School of Basic Medical Sciences, Central South University, Changsha, China
| |
Collapse
|
5
|
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
|
6
|
Zhang X, Zhang H, Li S, Fang F, Yin Y, Wang Q. Recent progresses in gut microbiome mediates obstructive sleep apnea-induced cardiovascular diseases. FASEB Bioadv 2024; 6:118-130. [PMID: 38585431 PMCID: PMC10995711 DOI: 10.1096/fba.2023-00153] [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: 12/12/2023] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 04/09/2024] Open
Abstract
Obstructive sleep apnea (OSA) is a multifactorial sleep disorder with a high prevalence in the general population. OSA is associated with an increased risk of developing cardiovascular diseases (CVDs), particularly hypertension, and is linked to worse outcomes. Although the correlation between OSA and CVDs is firmly established, the mechanisms are poorly understood. Continuous positive airway pressure is primary treatment for OSA reducing cardiovascular risk effectively, while is limited by inadequate compliance. Moreover, alternative treatments for cardiovascular complications in OSA are currently not available. Recently, there has been considerable attention on the significant correlation between gut microbiome and pathophysiological changes in OSA. Furthermore, gut microbiome has a significant impact on the cardiovascular complications that arise from OSA. Nevertheless, a detailed understanding of this association is lacking. This review examines recent advancements to clarify the link between the gut microbiome, OSA, and OSA-related CVDs, with a specific focus on hypertension, and also explores potential health advantages of adjuvant therapy that targets the gut microbiome in OSA.
Collapse
Affiliation(s)
- Xiaotong Zhang
- Shanxi Provincial People’s HospitalThe Fifth Clinical Medical College of Shanxi Medical UniversityTaiyuanChina
| | - Haifen Zhang
- Shanxi Provincial People’s HospitalThe Fifth Clinical Medical College of Shanxi Medical UniversityTaiyuanChina
| | - Shuai Li
- Shanxi Provincial People’s HospitalThe Fifth Clinical Medical College of Shanxi Medical UniversityTaiyuanChina
| | - Fan Fang
- Shanxi Provincial People’s HospitalThe Fifth Clinical Medical College of Shanxi Medical UniversityTaiyuanChina
| | - Yanran Yin
- Shanxi Provincial People’s HospitalThe Fifth Clinical Medical College of Shanxi Medical UniversityTaiyuanChina
| | - Qiang Wang
- Department of Infectious Disease, Shanxi Provincial People's HospitalThe Fifth Clinical Medical College of Shanxi Medical UniversityTaiyuanChina
| |
Collapse
|
7
|
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
|
8
|
Moravejolahkami AR, Shakibaei M, Fairley AM, Sharma M. Probiotics, prebiotics, and synbiotics in type 1 diabetes mellitus: A systematic review and meta-analysis of clinical trials. Diabetes Metab Res Rev 2024; 40:e3655. [PMID: 37183580 DOI: 10.1002/dmrr.3655] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/27/2023] [Accepted: 04/10/2023] [Indexed: 05/16/2023]
Abstract
Dysbiosis or imbalance of microbes in the gut has been associated with susceptibility and progression of type 1 diabetes mellitus (T1DM). The present systematic review and meta-analysis examined the effects of probiotics, prebiotics, and synbiotics on fasting blood glucose (FBG), haemoglobin A1c (HbA1c), C-peptide, and insulin requirements in T1DM patients. A systematic search for trials published up to October 2022 was conducted in PubMed, EMBASE, Scopus, Google Scholar, ScienceDirect, Web of Science, and the Central Cochrane Library. Random effect models were used to synthesise quantitative data by STATA14 . After the evaluation of 258 identified entries, five randomised controlled trials (n = 356; mean age = 11.7 years old) were included. The pooled effect size showed that FBG decreased following probiotic supplementation (weighted mean difference = -31.24 mg/dL; 95% confidence interval = -45.65, -16.83; p < 0.001), however, there was no significant improvement in serum HbA1c, C-peptide, and insulin requirements. Probiotic supplementation could be a complementary therapeutic strategy in T1DM. The evidence is limited; therefore, it is crucial to conduct more trials.
Collapse
Affiliation(s)
- Amir Reza Moravejolahkami
- Department of Clinical Nutrition, School of Nutrition & Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehdi Shakibaei
- Faculty of Medicine, Institute of Anatomy, Musculoskeletal & Tumor Biology Research Group, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Andrea Mary Fairley
- School of Biomedical, Nutritional and Sport Sciences, Newcastle University, Newcastle-Upon-Tyne, UK
| | - Manoj Sharma
- Department of Social & Behavioral Health, School of Public Health, University of Nevada, Las Vegas, Nevada, USA
- Department of Internal Medicine, Kirk Kerkorian School of Medicine, University of Nevada, Las Vegas, Nevada, USA
| |
Collapse
|
9
|
Salo TEI, Niinistö S, Korhonen TE, Pastell H, Reinivuo H, Takkinen HM, Ilonen J, Toppari J, Knip M, Veijola R, Virtanen SM. Intake and sources of dietary fibre and dietary fibre fractions in Finnish children. Br J Nutr 2023; 130:1416-1426. [PMID: 36803617 PMCID: PMC10511677 DOI: 10.1017/s0007114523000466] [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: 06/23/2022] [Revised: 02/01/2023] [Accepted: 02/13/2023] [Indexed: 02/23/2023]
Abstract
The current definition of dietary fibre was adopted by the Codex Alimentarius Commission in 2009, but implementation requires updating food composition databases with values based on appropriate analysis methods. Previous data on population intakes of dietary fibre fractions are sparse. We studied the intake and sources of total dietary fibre (TDF) and dietary fibre fractions insoluble dietary fibre (IDF), dietary fibre soluble in water but insoluble in 76 % aqueous ethanol (SDFP) and dietary fibre soluble in water and soluble in 76 % aqueous ethanol (SDFS) in Finnish children based on new CODEX-compliant values of the Finnish National Food Composition Database Fineli. Our sample included 5193 children at increased genetic risk of type 1 diabetes from the Type 1 Diabetes Prediction and Prevention birth cohort, born between 1996 and 2004. We assessed the intake and sources based on 3-day food records collected at the ages of 6 months, 1, 3 and 6 years. Both absolute and energy-adjusted intakes of TDF were associated with age, sex and breast-feeding status of the child. Children of older parents, parents with a higher level of education, non-smoking mothers and children with no older siblings had higher energy-adjusted TDF intake. IDF was the major dietary fibre fraction in non-breastfed children, followed by SDFP and SDFS. Cereal products, fruits and berries, potatoes and vegetables were major food sources of dietary fibre. Breast milk was a major source of dietary fibre in 6-month-olds due to its human milk oligosaccharide content and resulted in high SDFS intakes in breastfed children.
Collapse
Affiliation(s)
- Tuuli E. I. Salo
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, FI-00271 Helsinki, Finland
- Unit of Health Sciences, Faculty of Social Sciences, Tampere University, FI-33014 Tampere, Finland
| | - Sari Niinistö
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, FI-00271 Helsinki, Finland
| | - Tuuli E. Korhonen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, FI-00271 Helsinki, Finland
| | - Helena Pastell
- Finnish Food Authority, Mustialankatu 3, FI-00790 Helsinki, Finland
| | - Heli Reinivuo
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, FI-00271 Helsinki, Finland
| | - Hanna-Mari Takkinen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, FI-00271 Helsinki, Finland
- Unit of Health Sciences, Faculty of Social Sciences, Tampere University, FI-33014 Tampere, Finland
- Research, Development and Innovation Center, Tampere University Hospital, P.O. Box 2000, FI-33521 Tampere, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, FI-20014 Turku, Finland
| | - Jorma Toppari
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, FI-20520 Turku, Finland
- Department of Pediatrics, Turku University Hospital, FI-20520 Turku, Finland
| | - Mikael Knip
- Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, FI-00029 Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FI-00014 Helsinki, Finland
- Department of Pediatrics, Tampere University Hospital, FI-33521 Tampere, Finland
| | - Riitta Veijola
- Department of Pediatrics, PEDEGO Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, FI-90014 Oulu, Finland
- Department of Children and Adolescents, Oulu University Hospital, P.O. Box 10, FI-90029 Oulu, Finland
| | - Suvi M. Virtanen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, FI-00271 Helsinki, Finland
- Unit of Health Sciences, Faculty of Social Sciences, Tampere University, FI-33014 Tampere, Finland
- Research, Development and Innovation Center, Tampere University Hospital, P.O. Box 2000, FI-33521 Tampere, Finland
- Center for Child Health Research, Tampere University and Tampere University Hospital, FI-33014 Tampere, Finland
| |
Collapse
|
10
|
Hafezi SG, Seifi N, Bahari H, Mohammadi M, Ghasemabadi A, ferns GA, Farkhani EM, Ghayour-mobarhan M. The association between macronutrient intakes and coronavirus disease 2019 (COVID-19) in an Iranian population: applying a dynamical system model. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2023; 42:114. [PMID: 37884984 PMCID: PMC10601229 DOI: 10.1186/s41043-023-00448-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 09/22/2023] [Indexed: 10/28/2023]
Abstract
AIMS The possible role of lifestyle including diet on immunity led us to investigate the association between dietary macronutrient intake and COVID-19 in an Iranian population. METHODS Dietary intakes were recorded in the first phase of the MASHAD cohort study (started in 2007), using a 24-h dietary recall. To determine the COVID-19 incidence, data from all PCR-positive patients in Mashhad were recorded between February 2020 and June 2022. Dietary macronutrients were included in the regression model, adjusting for age and sex. System dynamical models were also applied. RESULTS The analysis included 1957 participants, including 193 COVID-19-positive patients. Dietary intakes of non-starch polysaccharides (NSP) and fiber were significantly lower in COVID-19 patients (P < 0.05). After adjusting for age and sex, starch and total sugar were significantly associated with COVID-19 infection ((OR = 1.0008, P = 0.001) and (OR = 1.0006, P = 0.026), respectively). There was also a significant association between dietary fiber intake and hospitalization (OR = 0.99, P = 0.018). In the dynamical system models, dietary intakes of cholesterol, polyunsaturated fatty acids (PUFA), and total sugar above 180.2 mg, 13.11 g, and 79.53 mg, respectively, were associated with an increased susceptibility to COVID-19 infection, while dietary fiber had a protective role. CONCLUSION Dietary intake of starch and total sugars was associated with increased odds of COVID-19, while fiber intake decreased the odds of hospitalization due to COVID-19. The dynamical system models showed that dietary intake of cholesterol, PUFAs, and total sugar was associated with an increased risk of COVID-19, while fiber had a protective role.
Collapse
Affiliation(s)
- Somayeh Ghiasi Hafezi
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Najmeh Seifi
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Bahari
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Mohammadi
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Gordon A. ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex UK
| | | | - Majid Ghayour-mobarhan
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
11
|
Plocica J, Guo F, Das JK, Kobayashi KS, Ficht TA, Alaniz RC, Song J, de Figueiredo P. Engineering live attenuated vaccines: Old dogs learning new tricks. J Transl Autoimmun 2023; 6:100198. [PMID: 37090898 PMCID: PMC10113845 DOI: 10.1016/j.jtauto.2023.100198] [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: 02/17/2023] [Revised: 03/24/2023] [Accepted: 03/26/2023] [Indexed: 03/29/2023] Open
Abstract
Autoimmune diseases such as rheumatoid arthritis and type 1 diabetes are increasingly common global problems. Concerns about increases in the prevalence of such diseases and the limited efficacy of conventional treatment regimens necessitates new therapies to address these challenges. Autoimmune disease severity and dysbiosis are interconnected. Although probiotics have been established as a therapy to rebalance the microbiome and suppress autoimmune symptoms, these microbes tend to lack a number of advantageous qualities found in non-commensal bacteria. Through attenuation and genetic manipulation, these non-commensal bacteria have been engineered into recombinant forms that offer malleable platforms capable of addressing the immune imbalances found in RA and T1D. Such bacteria have been engineered to express valuable gene products known to suppress autoimmunity such as anti-inflammatory cytokines, autoantigens, and enzymes synthesizing microbial metabolites. This review will highlight current and emerging trends in the field and discuss how they may be used to prevent and control autoimmune diseases.
Collapse
Affiliation(s)
- Julia Plocica
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Fengguang Guo
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Jugal Kishore Das
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Koichi S. Kobayashi
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
- Department of Immunology, Graduate School of Medicine, Hokkaido University Kita 15, Nishi 7, Kita-ku, Sapporo, 060-8638, Japan
- Institute of Vaccine Research and Development, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Thomas A. Ficht
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, 77845, USA
| | - Robert C. Alaniz
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Jianxun Song
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Paul de Figueiredo
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, 77845, USA
| |
Collapse
|
12
|
Piccioni A, Rosa F, Mannucci S, Manca F, Merra G, Chiloiro S, Candelli M, Covino M, Gasbarrini A, Franceschi F. Gut Microbiota, LADA, and Type 1 Diabetes Mellitus: An Evolving Relationship. Biomedicines 2023; 11:707. [PMID: 36979685 PMCID: PMC10045633 DOI: 10.3390/biomedicines11030707] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 02/14/2023] [Accepted: 02/21/2023] [Indexed: 03/03/2023] Open
Abstract
There is much evidence confirming the crucial role played by the gut microbiota in modulating the immune system in the onset of autoimmune diseases. In this article, we focus on the relationship between alterations in the microbiome and the onset of diabetes mellitus type 1 and LADA, in light of the latest evidence. We will then look at both how the role of the gut microbiota appears to be increasingly crucial in the pathogenesis of these disorders and how this aspect may be instrumental in the development of new potential therapeutic strategies that modulate the gut microbiota, such as probiotics, prebiotics, and fecal microbiota transplantation.
Collapse
Affiliation(s)
- Andrea Piccioni
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Federico Rosa
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Sergio Mannucci
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Federica Manca
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Giuseppe Merra
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy
| | - Sabrina Chiloiro
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Marcello Candelli
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Marcello Covino
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesco Franceschi
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
13
|
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
|
14
|
Wang L, Lian J, Zheng Q, Wang L, Wang Y, Yang D. Composition analysis and prebiotics properties of polysaccharides extracted from Lepista sordida submerged cultivation mycelium. Front Microbiol 2023; 13:1077322. [PMID: 36713178 PMCID: PMC9879602 DOI: 10.3389/fmicb.2022.1077322] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 12/15/2022] [Indexed: 01/15/2023] Open
Abstract
In this paper, Lepista sordida polysaccharides (LSP) were separated from Lepista sordida (L. sordida) mainly using the Ultrasonic-Micro Wave Synergy Extraction (UMSE) method and purified by graded alcohol precipitation. Three polysaccharide components: 40%-LSP-UMSE, 60%-LSP-UMSE, and 80%-LSP-UMSE were obtained and further analyzed the physicochemical properties, structural characteristics, and antioxidant activity. And the effects on the proliferation of Lactobacillus casei of three polysaccharide components were studied. The characteristic absorption peaks and the β-glycosidic bond of three polysaccharide components were the direct expression at UV 200 nm using UV and FT-IR spectroscopy. The three polysaccharide components were mainly composed of glucose, mannose, galactose, and ribose using high-performance liquid chromatography (HPLC) analysis. The antioxidant activity study revealed that the polysaccharides obtained by the UMSE method had better antioxidant activity compared to the traditional "Hot Water Extraction (HWE)" method. In addition, the polysaccharide components promoted the proliferation of L. casei to some extent. 40%-LSP-UMSE, 80%-LSP-UMSE as the carbon source had better acid production than the control inulin. Three LSP-UMSE used as a carbon source compared with glucose for culturing L. casei could significantly improve its tolerance to bile salts. Results are helpful to develop the bioactive polysaccharides from Lepista sordida and beneficial to develop a unique health and functional product in the future.
Collapse
|
15
|
Shabani-Mirzaee H, Haghshenas Z, Malekiantaghi A, Vigeh M, Mahdavi F, Eftekhari K. The effect of oral probiotics on glycated haemoglobin levels in children with type 1 diabetes mellitus - a randomized clinical trial. Pediatr Endocrinol Diabetes Metab 2023; 29:128-133. [PMID: 38031828 PMCID: PMC10679923 DOI: 10.5114/pedm.2023.132025] [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: 11/18/2022] [Accepted: 06/08/2023] [Indexed: 12/01/2023]
Abstract
INTRODUCTION Proper control of blood glucose in children with type 1 diabetes has a direct effect on their metabolism and quality of life by reducing the risk of complications. The use of probiotics may have a beneficial effect on glucose levels. PURPOSE The aim of this study was to evaluate the effect of oral consumption of probiotics on glycosylated haemoglobin in children with type 1 diabetes. MATERIAL AND METHODS In this single-blind randomized controlled clinical trial, 52 children with type 1 diabetes were studied. We created 2 groups of 26 individuals each. The probiotic group received a daily probiotic capsule for 90 days, in addition to routine insulin therapy. The control group received only insulin therapy. Blood samples were taken to measure HbA1c, fasting plasma glucose, and lipid profiles at the beginning and end of the trial. RESULTS The study showed that HbA1c was high in both groups, but this increase was lower in the probiotic group than in the control group. This difference was not statistically significant. The mean level of fasting plasma glucose in the probiotic group was significantly reduced compared to the control group (p = 0.016). CONCLUSIONS According to the results of our study, consumption of oral probiotics has no significant effect on HbA1c levels in children with type 1 diabetes mellitus.
Collapse
Affiliation(s)
- Hosein Shabani-Mirzaee
- Department of Pediatric Endocrinology, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Haghshenas
- Department of Pediatric Endocrinology, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mohsen Vigeh
- Maternal-Fetal Medicine Research Center, Tehran University of Medical Sciences, Iran
| | - Fazeleh Mahdavi
- Department of Pediatric Endocrinology, Tehran University of Medical Sciences, Tehran, Iran
| | - Kambiz Eftekhari
- Pediatrics, Tehran University of Medical Sciences, Iran
- Pediatric Gastroenterology and Hepatology Research Center, Tehran University of Medical Sciences, Iran
| |
Collapse
|
16
|
Functional and Taxonomic Traits of the Gut Microbiota in Type 1 Diabetes Children at the Onset: A Metaproteomic Study. Int J Mol Sci 2022; 23:ijms232415982. [PMID: 36555624 PMCID: PMC9787575 DOI: 10.3390/ijms232415982] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/02/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune metabolic disorder with onset in pediatric/adolescent age, characterized by insufficient insulin production, due to a progressive destruction of pancreatic β-cells. Evidence on the correlation between the human gut microbiota (GM) composition and T1D insurgence has been recently reported. In particular, 16S rRNA-based metagenomics has been intensively employed in the last decade in a number of investigations focused on GM representation in relation to a pre-disease state or to a response to clinical treatments. On the other hand, few works have been published using alternative functional omics, which is more suitable to provide a different interpretation of such a relationship. In this work, we pursued a comprehensive metaproteomic investigation on T1D children compared with a group of siblings (SIBL) and a reference control group (CTRL) composed of aged matched healthy subjects, with the aim of finding features in the T1D patients' GM to be related with the onset of the disease. Modulated metaproteins were found either by comparing T1D with CTRL and SIBL or by stratifying T1D by insulin need (IN), as a proxy of β-cells damage, showing some functional and taxonomic traits of the GM, possibly related to the disease onset at different stages of severity.
Collapse
|
17
|
Pavlidou E, Fasoulas A, Mantzorou M, Giaginis C. Clinical Evidence on the Potential Beneficial Effects of Probiotics and Prebiotics in Cardiovascular Disease. Int J Mol Sci 2022; 23:ijms232415898. [PMID: 36555535 PMCID: PMC9779729 DOI: 10.3390/ijms232415898] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/08/2022] [Accepted: 12/11/2022] [Indexed: 12/15/2022] Open
Abstract
The 'gut microbiome'-the hundreds of trillions of bacteria in the human gastrointestinal tract-serves several functions. The gut microbiome includes all the microorganisms, bacteria, viruses, protozoa, and fungi in the gastrointestinal tract and their genetic material. It helps digest indigestible foods and produces nutrients. Through the metabolism of sugars and proteins, it helps the intestinal barrier, the immune system, and metabolism. Some bacteria, such as those in the gut microbiome, cause disease, but others are essential to our health. These "good" microbes protect us from pathogens. Numerous studies have linked an unhealthy gut microbiome to obesity, insulin resistance, depression, and cardiometabolic risk factors. To maximize probiotic benefits in each case, knowledge of probiotic bacterial strains and how to consume them should be increased. This study aims to examine the benefits of probiotic and prebiotic organisms on cardiovascular health, specifically on heart disease, coronary heart disease, stroke, and hypertension. To complete the research, a literature review was conducted by gathering clinical studies and data. The clinical evidence demonstrates the beneficial effect of probiotics and prebiotic microorganisms on the gut microbiome, which has multiple benefits for overall health and especially for cardiovascular diseases.
Collapse
|
18
|
Choneva M, Shishmanova-Doseva M, Dimov I, Boyanov K, Dimitrov I, Vlaykova T, Georgieva K, Hrischev P, Bivolarska A. Xylooligosaccharides and aerobic training regulate metabolism and behavior in rats with streptozotocin-induced type 1 diabetes. Open Med (Wars) 2022; 17:1632-1644. [PMID: 36329786 PMCID: PMC9579861 DOI: 10.1515/med-2022-0579] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 09/06/2022] [Accepted: 09/09/2022] [Indexed: 11/15/2022] Open
Abstract
Type 1 diabetes mellitus is characterized with decreased microbial diversity. Gut microbiota is essential for the normal physiological functioning of many organs, especially the brain. Prebiotics are selectively fermentable oligosaccharides [xylooligosaccharides (XOS), galactooligosaccharides, etc.] that promote the growth and activity of gut microbes and influence the gut-brain axis. Aerobic exercise is a non-pharmacological approach for the control of diabetes and could improve cognitive functions. The potential beneficial effect of XOS and/or aerobic training on cognition, the lipid profile and oxidative stress markers of experimental rats were evaluated in this study. Male Wistar rats were randomly divided into three streptozotocin-induced diabetic groups and a control group. Some of the rats, either on a XOS treatment or a standard diet, underwent aerobic training. The results showed that the aerobic training independently lowered the total cholesterol levels compared to the sedentary diabetic rats (p = 0.032), while XOS lowers the malondialdehyde levels in the trained diabetic rats (p = 0.034). What is more the exercise, independently or in combination with XOS beneficially affected all parameters of the behavioral tests. We conclude that aerobic exercises alone or in a combination with the prebiotic XOS could ameliorate the dyslipidemia, oxidative stress, and cognitive abilities in experimental type 1 diabetic animals.
Collapse
Affiliation(s)
- Mariya Choneva
- Department of Medical Biochemistry, Faculty of Pharmacy, Medical University of Plovdiv, 15 A, Vassil Aprilov Blvd., Plovdiv, 4002, Bulgaria
| | - Michaela Shishmanova-Doseva
- Department of Pharmacology, Toxicology and Pharmacotherapy, Faculty of Pharmacy, Medical University of Plovdiv, 15 A, Vassil Aprilov Blvd., Plovdiv, 4002, Bulgaria
| | - Ivica Dimov
- Department of Medical Biochemistry, Faculty of Pharmacy, Medical University of Plovdiv, 15 A, Vassil Aprilov Blvd., Plovdiv, 4002, Bulgaria
| | - Krasimir Boyanov
- Department of Medical Biochemistry, Faculty of Pharmacy, Medical University of Plovdiv, 15 A, Vassil Aprilov Blvd., Plovdiv, 4002, Bulgaria
| | - Iliyan Dimitrov
- Department of Medical Biochemistry, Faculty of Pharmacy, Medical University of Plovdiv, 15 A, Vassil Aprilov Blvd., Plovdiv, 4002, Bulgaria
| | - Tatyana Vlaykova
- Department of Medical Biochemistry, Faculty of Pharmacy, Medical University of Plovdiv, 15 A, Vassil Aprilov Blvd., Plovdiv, 4002, Bulgaria
| | - Katerina Georgieva
- Department of Physiology, Faculty of Pharmacy, Medical University of Plovdiv, 15 A, Vassil Aprilov Blvd., Plovdiv, 4002, Bulgaria
| | - Petar Hrischev
- Department of Physiology, Faculty of Pharmacy, Medical University of Plovdiv, 15 A, Vassil Aprilov Blvd., Plovdiv, 4002, Bulgaria
| | - Anelia Bivolarska
- Department of Medical Biochemistry, Faculty of Pharmacy, Medical University of Plovdiv, 15 A, Vassil Aprilov Blvd., Plovdiv, 4002, Bulgaria
| |
Collapse
|
19
|
Bucheli JEV, Todorov SD, Holzapfel WH. Role of gastrointestinal microbial populations, a terra incognita of the human body in the management of intestinal bowel disease and metabolic disorders. Benef Microbes 2022; 13:295-318. [PMID: 35866598 DOI: 10.3920/bm2022.0022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Intestinal bowel disease (IBD) is a chronic immune-mediated clinical condition that affects the gastrointestinal tract and is mediated by an inflammatory response. Although it has been extensively studied, the multifactorial aetiology of this disorder makes it difficult to fully understand all the involved mechanisms in its development and therefore its treatment. In recent years, the fundamental role played by the human microbiota in the pathogenesis of IBD has been emphasised. Microbial imbalances in the gut bacterial communities and a lower species diversity in patients suffering from inflammatory gastrointestinal disorders compared to healthy individuals have been reported as principal factors in the development of IBD. These served to support scientific arguments for the use of probiotic microorganisms in alternative approaches for the prevention and treatment of IBD. In a homeostatic environment, the presence of bacteria (including probiotics) on the intestinal epithelial surface activates a cascade of processes by which immune responses inhibited and thereby commensal organisms maintained. At the same time these processes may support activities against specific pathogenic bacteria. In dysbiosis, these underlying mechanisms will serve to provoke a proinflammatory response, that, in combination with the use of antibiotics and the genetic predisposition of the host, will culminate in the development of IBD. In this review, we summarised the main causes of IBD, the physiological mechanisms involved and the related bacterial groups most frequently associated with these processes. The intention was to enable a better understanding of the interaction between the intestinal microbiota and the host, and to suggest possibilities by which this knowledge can be useful for the development of new therapeutic treatments.
Collapse
Affiliation(s)
- J E Vazquez Bucheli
- Human Effective Microbes, Department of Advanced Convergence, Handong Global University, Pohang, Gyeongbuk 37554, Republic of Korea
| | - S D Todorov
- ProBacLab, Department of Advanced Convergence, Handong Global University, Pohang, Gyeongbuk 37554, Republic of Korea
| | - W H Holzapfel
- Human Effective Microbes, Department of Advanced Convergence, Handong Global University, Pohang, Gyeongbuk 37554, Republic of Korea
| |
Collapse
|
20
|
Zemanova N, Omelka R, Mondockova V, Kovacova V, Martiniakova M. Roles of Gut Microbiome in Bone Homeostasis and Its Relationship with Bone-Related Diseases. BIOLOGY 2022; 11:1402. [PMID: 36290306 PMCID: PMC9598716 DOI: 10.3390/biology11101402] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/14/2022] [Accepted: 09/19/2022] [Indexed: 11/16/2022]
Abstract
The extended microbial genome-the gut microbiome (GM)-plays a significant role in host health and disease. It is able to influence a number of physiological functions. During dysbiosis, GM is associated with the development of various chronic diseases with impaired bone quality. In general, GM is important for bone homeostasis and can affect it via several mechanisms. This review describes the roles of GM in bone homeostasis through influencing the immune and endocrine functions, short-chain fatty acids production, calcium absorption and the gut-brain axis. The relationship between GM composition and several bone-related diseases, specifically osteoporosis, osteoarthritis, rheumatoid arthritis, diabetes mellitus, obesity and bone cancer, is also highlighted and summarized. GM manipulation may become a future adjuvant therapy in the prevention of many chronic diseases. Therefore, the beneficial effects of probiotic therapy to improve the health status of individuals with aforementioned diseases are provided, but further studies are needed to clearly confirm its effectiveness. Recent evidence suggests that GM is responsible for direct and indirect effects on drug efficacy. Accordingly, various GM alterations and interactions related to the treatment of bone-related diseases are mentioned as well.
Collapse
Affiliation(s)
- Nina Zemanova
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 74 Nitra, Slovakia
| | - Radoslav Omelka
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 74 Nitra, Slovakia
| | - Vladimira Mondockova
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 74 Nitra, Slovakia
| | - Veronika Kovacova
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 74 Nitra, Slovakia
| | - Monika Martiniakova
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 74 Nitra, Slovakia
| |
Collapse
|
21
|
Webb-Robertson BJM, Nakayasu ES, Frohnert BI, Bramer LM, Akers SM, Norris JM, Vehik K, Ziegler AG, Metz TO, Rich SS, Rewers MJ. Integration of Infant Metabolite, Genetic, and Islet Autoimmunity Signatures to Predict Type 1 Diabetes by Age 6 Years. J Clin Endocrinol Metab 2022; 107:2329-2338. [PMID: 35468213 PMCID: PMC9282254 DOI: 10.1210/clinem/dgac225] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Indexed: 02/08/2023]
Abstract
CONTEXT Biomarkers that can accurately predict risk of type 1 diabetes (T1D) in genetically predisposed children can facilitate interventions to delay or prevent the disease. OBJECTIVE This work aimed to determine if a combination of genetic, immunologic, and metabolic features, measured at infancy, can be used to predict the likelihood that a child will develop T1D by age 6 years. METHODS Newborns with human leukocyte antigen (HLA) typing were enrolled in the prospective birth cohort of The Environmental Determinants of Diabetes in the Young (TEDDY). TEDDY ascertained children in Finland, Germany, Sweden, and the United States. TEDDY children were either from the general population or from families with T1D with an HLA genotype associated with T1D specific to TEDDY eligibility criteria. From the TEDDY cohort there were 702 children will all data sources measured at ages 3, 6, and 9 months, 11.4% of whom progressed to T1D by age 6 years. The main outcome measure was a diagnosis of T1D as diagnosed by American Diabetes Association criteria. RESULTS Machine learning-based feature selection yielded classifiers based on disparate demographic, immunologic, genetic, and metabolite features. The accuracy of the model using all available data evaluated by the area under a receiver operating characteristic curve is 0.84. Reducing to only 3- and 9-month measurements did not reduce the area under the curve significantly. Metabolomics had the largest value when evaluating the accuracy at a low false-positive rate. CONCLUSION The metabolite features identified as important for progression to T1D by age 6 years point to altered sugar metabolism in infancy. Integrating this information with classic risk factors improves prediction of the progression to T1D in early childhood.
Collapse
Affiliation(s)
- Bobbie-Jo M Webb-Robertson
- Correspondence: Bobbie-Jo Webb-Robertson, PhD, Biological Sciences Division, Pacific Northwest National Laboratory, 902 Battelle Blvd, MSIN: J4-18, Richland, WA 99352, USA.
| | - Ernesto S Nakayasu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352,USA
| | - Brigitte I Frohnert
- Barbara Davis Center for Childhood Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Lisa M Bramer
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352,USA
| | - Sarah M Akers
- Computing & Analytics Division, Pacific Northwest National Laboratory, Richland, Washington 99352, USA
| | - Jill M Norris
- Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Kendra Vehik
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612, USA
| | - Anette-G Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Kilinikum rechts der Isar, Technische Universität München, 80333 Munich, Germany
- Forschergruppe Diabetes e.V., 85764 Neuherberg, Germany
| | - Thomas O Metz
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352,USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia 22908,USA
| | - Marian J Rewers
- Barbara Davis Center for Childhood Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| |
Collapse
|
22
|
The Activity of Prebiotics and Probiotics in Hepatogastrointestinal Disorders and Diseases Associated with Metabolic Syndrome. Int J Mol Sci 2022; 23:ijms23137229. [PMID: 35806234 PMCID: PMC9266451 DOI: 10.3390/ijms23137229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 06/27/2022] [Indexed: 12/11/2022] Open
Abstract
The components of metabolic syndrome (MetS) and hepatogastrointestinal diseases are widespread worldwide, since many factors associated with lifestyle and diet influence their development and correlation. Due to these growing health problems, it is necessary to search for effective alternatives for prevention or adjuvants in treating them. The positive impact of regulated microbiota on health is known; however, states of dysbiosis are closely related to the development of the conditions mentioned above. Therefore, the role of prebiotics, probiotics, or symbiotic complexes has been extensively evaluated; the results are favorable, showing that they play a crucial role in the regulation of the immune system, the metabolism of carbohydrates and lipids, and the biotransformation of bile acids, as well as the modulation of their central receptors FXR and TGR-5, which also have essential immunomodulatory and metabolic activities. It has also been observed that they can benefit the host by displacing pathogenic species, improving the dysbiosis state in MetS. Current studies have reported that paraprobiotics (dead or inactive probiotics) or postbiotics (metabolites generated by active probiotics) also benefit hepatogastrointestinal health.
Collapse
|
23
|
Effects of Butyrate Supplementation on Inflammation and Kidney Parameters in Type 1 Diabetes: A Randomized, Double-Blind, Placebo-Controlled Trial. J Clin Med 2022; 11:jcm11133573. [PMID: 35806857 PMCID: PMC9267418 DOI: 10.3390/jcm11133573] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 04/27/2022] [Accepted: 06/09/2022] [Indexed: 12/19/2022] Open
Abstract
Type 1 diabetes is associated with increased intestinal inflammation and decreased abundance of butyrate-producing bacteria. We investigated the effect of butyrate on inflammation, kidney parameters, HbA1c, serum metabolites and gastrointestinal symptoms in persons with type 1 diabetes, albuminuria and intestinal inflammation. We conducted a randomized placebo-controlled, double-blind, parallel clinical study involving 53 participants randomized to 3.6 g sodium butyrate daily or placebo for 12 weeks. The primary endpoint was the change in fecal calprotectin. Additional endpoints were the change in fecal short chain fatty acids, intestinal alkaline phosphatase activity and immunoglobulins, serum lipopolysaccharide, CRP, albuminuria, kidney function, HbA1c, metabolites and gastrointestinal symptoms. The mean age was 54 ± 13 years, and the median [Q1:Q3] urinary albumin excretion was 46 [14:121] mg/g. The median fecal calprotectin in the butyrate group was 48 [26:100] μg/g at baseline, and the change was −1.0 [−20:10] μg/g; the median in the placebo group was 61 [25:139] μg/g at baseline, and the change was −12 [−95:1] μg/g. The difference between the groups was not significant (p = 0.24); neither did we find an effect of butyrate compared to placebo on the other inflammatory markers, kidney parameters, HbA1c, metabolites nor gastrointestinal symptoms. Twelve weeks of butyrate supplementation did not reduce intestinal inflammation in persons with type 1 diabetes, albuminuria and intestinal inflammation.
Collapse
|
24
|
Winiarska-Mieczan A, Tomaszewska E, Donaldson J, Jachimowicz K. The Role of Nutritional Factors in the Modulation of the Composition of the Gut Microbiota in People with Autoimmune Diabetes. Nutrients 2022; 14:2498. [PMID: 35745227 PMCID: PMC9227140 DOI: 10.3390/nu14122498] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 12/14/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is a disease marked by oxidative stress, chronic inflammation, and the presence of autoantibodies. The gut microbiota has been shown to be involved in the alleviation of oxidative stress and inflammation as well as strengthening immunity, thus its' possible involvement in the pathogenesis of T1DM has been highlighted. The goal of the present study is to analyze information on the relationship between the structure of the intestinal microbiome and the occurrence of T1DM. The modification of the intestinal microbiota can increase the proportion of SCFA-producing bacteria, which could in turn be effective in the prevention and/or treatment of T1DM. The increased daily intake of soluble and non-soluble fibers, as well as the inclusion of pro-biotics, prebiotics, herbs, spices, and teas that are sources of phytobiotics, in the diet, could be important in improving the composition and activity of the microbiota and thus in the prevention of metabolic disorders. Understanding how the microbiota interacts with immune cells to create immune tolerance could enable the development of new therapeutic strategies for T1DM and improve the quality of life of people with T1DM.
Collapse
Affiliation(s)
- Anna Winiarska-Mieczan
- Department of Bromatology and Nutrition Physiology, Institute of Animal Nutrition and Bromatology, University of Life Sciences in Lublin, Akademicka St. 13, 20-950 Lublin, Poland;
| | - Ewa Tomaszewska
- Department of Animal Physiology, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Akademicka St. 12, 20-950 Lublin, Poland
| | - Janine Donaldson
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa;
| | - Karolina Jachimowicz
- Department of Bromatology and Nutrition Physiology, Institute of Animal Nutrition and Bromatology, University of Life Sciences in Lublin, Akademicka St. 13, 20-950 Lublin, Poland;
| |
Collapse
|
25
|
Tegegne BA, Kebede B. Probiotics, their prophylactic and therapeutic applications in human health development: A review of the literature. Heliyon 2022; 8:e09725. [PMID: 35785237 PMCID: PMC9240980 DOI: 10.1016/j.heliyon.2022.e09725] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/03/2022] [Accepted: 06/09/2022] [Indexed: 02/07/2023] Open
Abstract
Antibiotics do not differentiate between good and bad germs, disrupting normal microflora and causing vitamin deficiency in the human body. They also kill healthy bacteria in the gut and genital tract on a large scale, weakening the host's defense mechanism. Probiotics are a colony of bacteria that live in our intestines and are regarded as a metabolic 'organ' due to their beneficial effects on human health, including metabolism and immunological function. They are used in clinical settings to prevent and treat conditions such as diarrhoea, colon cancer, hypertension, diabetes, acute pancreatitis, Helicobacter pylori infection, ventilator-associated pneumonia, migraine and autism. Probiotics may modify immunological activity by increasing innate and adaptive immune responses, altering microbial habitat in the intestine, improving gut barrier function, competitive adherence to the mucosa and epithelium, and producing antimicrobial compounds. The aim of this study is to index that further in depth researches to be conducted on probiotics pivotal role in the prophylaxis and therapeutic usage for a variety of disease that may or may not have treatment alternatives. Key words such as probiotics, microbiota, prophylactics, and therapeutic applications were searched extensively in research databases such as PubMed, PubMed Central (PMC), Scopus, Web of Science, Research Gate, Google Scholar, and Cochrane Library. This concise narrative review article summarized primarily the history, selection, mechanism/mode of action, recent advances in prophylactic and therapeutic applications, and future directions in the use of probiotics for prophylactic and therapeutic applications.
Collapse
|
26
|
Environmental Factors and the Risk of Developing Type 1 Diabetes-Old Disease and New Data. BIOLOGY 2022; 11:biology11040608. [PMID: 35453807 PMCID: PMC9027552 DOI: 10.3390/biology11040608] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/10/2022] [Accepted: 04/14/2022] [Indexed: 12/16/2022]
Abstract
Simple Summary Despite many studies, the risk factors of type 1 diabetes (T1DM) in children and adolescents are still not fully understood and remain a big challenge. Therefore, an extensive online search for scientific research on factors related to diabetes has been performed for the identification of new factors of unexplained etiology. A better understanding of the role of viral, bacterial, and yeast-like fungi infections related to the risk of T1DM in children and adolescents and the identification of new risk factors, especially those spread by the droplet route, is of great importance for people and families with diabetes. Abstract The incidence of type 1 diabetes (T1D) is increasing worldwide. The onset of T1D usually occurs in childhood and is caused by the selective destruction of insulin-producing pancreatic islet cells (β-cells) by autoreactive T cells, leading to insulin deficiency. Despite advanced research and enormous progress in medicine, the causes of T1D are still not fully understood. Therefore, an extensive online search for scientific research on environmental factors associated with diabetes and the identification of new factors of unexplained etiology has been carried out using the PubMed, Cochrane, and Embase databases. The search results were limited to the past 11 years of research and discovered 143 manuscripts published between 2011 and 2022. Additionally, 21 manuscripts from between 2000 and 2010 and 3 manuscripts from 1974 to 2000 were referenced for historical reference as the first studies showcasing a certain phenomenon or mechanism. More and more scientists are inclined to believe that environmental factors are responsible for the increased incidence of diabetes. Research results show that higher T1D incidence is associated with vitamin D deficiency, a colder climate, and pollution of the environment, as well as the influence of viral, bacterial, and yeast-like fungi infections. The key viral infections affecting the risk of developing T1DM are rubella virus, mumps virus, Coxsackie virus, cytomegalovirus, and enterovirus. Since 2020, i.e., from the beginning of the COVID-19 pandemic, more and more studies have been looking for a link between Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) and diabetes development. A better understanding of the role of viral, bacterial, and yeast-like fungi infections related to the risk of T1DM in children and adolescents and the identification of new risk factors, especially those spread by the droplet route, is of great importance for people and families with diabetes.
Collapse
|
27
|
Liang W, Enée E, Andre-Vallee C, Falcone M, Sun J, Diana J. Intestinal Cathelicidin Antimicrobial Peptide Shapes a Protective Neonatal Gut Microbiota Against Pancreatic Autoimmunity. Gastroenterology 2022; 162:1288-1302.e16. [PMID: 34973295 DOI: 10.1053/j.gastro.2021.12.272] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 12/08/2021] [Accepted: 12/21/2021] [Indexed: 01/13/2023]
Abstract
BACKGROUND & AIMS Alteration of the gut microbiota is implicated in the development of autoimmune type 1 diabetes (T1D), as shown in humans and the nonobese diabetic (NOD) mouse model. However, how gut dysbiosis arises and promotes the autoimmune response remains an open question. We investigated whether early events affecting the intestinal homeostasis in newborn NOD mice may explain the development of the autoimmune response in the adult pancreas. METHODS We profiled the transcriptome and the microbiota in the colon between newborn NOD mice and nonautoimmune strains. We identified a seminal defect in the intestinal homeostasis of newborn NOD mice and deciphered the mechanism linking this defect to the diabetogenic response in the adult. RESULTS We determined that the cathelicidin-related antimicrobial peptide (CRAMP) expression was defective in the colon of newborn NOD mice, allowing inducing dysbiosis. Dysbiosis stimulated the colonic epithelial cells to produce type I interferons that pathologically imprinted the local neonatal immune system. This pathological immune imprinting later promoted the pancreatic autoimmune response in the adult and the development of diabetes. Increasing colonic CRAMP expression in newborn NOD mice by means of local CRAMP treatment or CRAMP-expressing probiotic restored colonic homeostasis and halted the diabetogenic response, preventing autoimmune diabetes. CONCLUSIONS We identified whether a defective colonic expression in the CRAMP antimicrobial peptide induces dysbiosis, contributing to autoimmunity in the pancreas. Hence, the manipulation of intestinal antimicrobial peptides may be considered a relevant therapeutic approach to prevent autoimmune diabetes in at-risk children.
Collapse
Affiliation(s)
- Wenjie Liang
- Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Université de Paris, Paris, France
| | - Emmanuelle Enée
- Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Université de Paris, Paris, France
| | - Cédric Andre-Vallee
- Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Université de Paris, Paris, France
| | - Marika Falcone
- Experimental Diabetes Unit, Division of Immunology, Transplantation and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico, San Raffaele Scientific Institute, Milan, Italy
| | - Jia Sun
- Nutritional Immunology and Translational Medicine Laboratory, State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, People's Republic of China.
| | - Julien Diana
- Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Université de Paris, Paris, France.
| |
Collapse
|
28
|
Bedi S, Richardson TM, Jia B, Saab H, Brinkman FSL, Westley M. Similarities between bacterial GAD and human GAD65: Implications in gut mediated autoimmune type 1 diabetes. PLoS One 2022; 17:e0261103. [PMID: 35196314 PMCID: PMC8865633 DOI: 10.1371/journal.pone.0261103] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/21/2022] [Indexed: 12/25/2022] Open
Abstract
A variety of islet autoantibodies (AAbs) can predict and possibly dictate eventual type 1 diabetes (T1D) diagnosis. Upwards of 75% of those with T1D are positive for AAbs against glutamic acid decarboxylase (GAD65 or GAD), a producer of gamma-aminobutyric acid (GABA) in human pancreatic beta cells. Interestingly, bacterial populations within the human gut also express GAD and produce GABA. Evidence suggests that dysbiosis of the microbiome may correlate with T1D pathogenesis and physiology. Therefore, autoimmune linkages between the gut microbiome and islets susceptible to autoimmune attack need to be further elucidated. Utilizing in silico analyses, we show that 25 GAD sequences from human gut bacterial sources show sequence and motif similarities to human beta cell GAD65. Our motif analyses determined that most gut GAD sequences contain the pyroxical dependent decarboxylase (PDD) domain of human GAD65, which is important for its enzymatic activity. Additionally, we showed overlap with known human GAD65 T cell receptor epitopes, which may implicate the immune destruction of beta cells. Thus, we propose a physiological hypothesis in which changes in the gut microbiome in those with T1D result in a release of bacterial GAD, thus causing miseducation of the host immune system. Due to the notable similarities we found between human and bacterial GAD, these deputized immune cells may then target human beta cells leading to the development of T1D.
Collapse
Affiliation(s)
- Suhana Bedi
- Department of Natural Sciences and Mathematics, The University of Texas at Dallas, Richardson, TX, United States of America
| | - Tiffany M. Richardson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States of America
| | - Baofeng Jia
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, CA, United States of America
| | - Hadeel Saab
- Intern, The(sugar)science, Los Angeles, CA, United States of America
| | - Fiona S. L. Brinkman
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, CA, United States of America
| | - Monica Westley
- Founder, The(sugar)science, Los Angeles, CA, United States of America
- * E-mail:
| |
Collapse
|
29
|
Evidence and possible mechanisms of probiotics in the management of type 1 diabetes mellitus. J Diabetes Metab Disord 2022; 21:1081-1094. [PMID: 35673472 PMCID: PMC9167374 DOI: 10.1007/s40200-022-01006-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 02/12/2022] [Indexed: 10/19/2022]
Abstract
Abstract Type 1 diabetes mellitus (T1DM) is one of the most common chronic immune-mediated diseases. The prevalence is worldwide especially among children and young adults. The destruction of the pancreatic β-cells due to some abnormalities in the immune system characterizes T1DM. Considering the high burden of the disease and its impact on human health, researchers have made great efforts during the last decades; investigating the disease pathogenesis and discovering new strategies for its management. Fortunately, probiotics have been found as potential remedies for T1DM. This review aims to explore the potentialities of probiotics in managing T1DM and its complications. Based on the outcomes of human and animal studies carried out from 2016 to 2021, the review hopes to assess the effectiveness of probiotics in the prevention and treatment of T1DM and its complications. We first tried to explain the disease's pathogenesis, and highlighted the possible mechanisms involved in these potentialities of probiotics. We concluded that, probiotics can be used as possible therapeutic tools for the management of T1DM. Possible mechanisms of action of probiotics include; the modulation of the gut microbiota, the regulation of inflammation-related cytokines, the production of short chain fatty acids (SCFAs), and the regulation of GLP-1. However, we recommend further studies especially human trials should be carried out to investigate these potentialities of probiotics. Highlights • T1DM is highly prevalent worldwide, causing high morbidity and mortality especially among children and young adults• Gut microbiota plays a significant role in the pathogenesis of T1DM via an interconnection with the immune system• Probiotics can be used as possible therapeutic tools for the management of T1DM• Possible mechanisms of action of probiotics include the modulation of the gut microbiota, the regulation of inflammation-related cytokines, the production of SCFAs, and the regulation of GLP-1.
Collapse
|
30
|
Wang CH, Yen HR, Lu WL, Ho HH, Lin WY, Kuo YW, Huang YY, Tsai SY, Lin HC. Adjuvant Probiotics of Lactobacillus salivarius subsp. salicinius AP-32, L. johnsonii MH-68, and Bifidobacterium animalis subsp. lactis CP-9 Attenuate Glycemic Levels and Inflammatory Cytokines in Patients With Type 1 Diabetes Mellitus. Front Endocrinol (Lausanne) 2022; 13:754401. [PMID: 35299968 PMCID: PMC8921459 DOI: 10.3389/fendo.2022.754401] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/31/2022] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Type 1 diabetes mellitus (T1DM) is characterized by autoimmune destruction of pancreatic β cells. Previous study has discovered that probiotic strains residing in the gut play essential roles in host immune regulation. However, few clinical results demonstrated probiotic would actually benefit in attenuating glycated hemoglobin (HbA1c) along with inflammatory cytokine levels of the T1DM patients and analyzed their gut microbiota profile at the same time. In this clinical trial, we evaluated the therapeutic efficacy of probiotics on HbA1c along with inflammatory cytokine levels of T1DM patients to determine an alternative administration mode for T1DM medication. The probiotics changed T1DM gut microbiota profile will be measured by next-generation sequencing (NGS). RESEARCH DESIGN AND METHODS A randomized, double-blind, placebo-controlled trial was performed at China Medical University Hospital. T1DM patients between 6 and 18 years of age were enrolled. 27 patients were administered regular insulin therapy plus capsules containing probiotic strains Lactobacillus salivarius subsp. salicinius AP-32, L. johnsonii MH-68, and Bifidobacterium animalis subsp. lactis CP-9 daily for 6 months, and 29 patients were administered insulin therapy without extra probiotic supplement as placebo group. The variations of fasting blood glucose and HbA1c in these patients were analyzed. In addition, serum levels of inflammatory cytokines and anti-inflammatory cytokine were assessed using enzyme-linked immunosorbent assay. Patients' stool microbiota were all subjects to NGS analysis. RESULTS NGS data showed elevated populations of Bifidobacterium animalis, Akkermansia muciniphila and Lactobacillus salivarius in the gut of patients with T1DM who were taking probiotics. Patients with T1DM who were administered probiotics showed significantly reduced fasting blood glucose levels compared with the before-intervention levels. The HbA1c levels of the patients also improved after administration of probiotics. The concentrations of IL-8, IL-17, MIP-1β, RANTES, and TNF-α were significantly reduced and were associated with an increased TGF-β1 expression after probiotic intervention. The persistence effect of glycemic control and immunomodulation were observed even 3 months after discontinuation of the probiotics. CONCLUSIONS Here, we found that conventional insulin therapy plus probiotics supplementation attenuated T1DM symptoms than receiving insulin treatment only. Probiotics supplementation with insulin treatment changed gut microbiota and revealed better outcome in stabilizing glycemic levels and reducing HbA1c levels in patients with T1DM through beneficial regulation of immune cytokines. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov, identifier NCT03880760.
Collapse
Affiliation(s)
- Chung-Hsing Wang
- Division of Medical Genetics, Pediatric Endocrinology & Metabolism, China Medical University Children’s Hospital, China Medical University, Taichung, Taiwan
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Hung-Rong Yen
- Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan
- Research Center for Traditional Chinese Medicine, China Medical University Hospital, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Wen-Li Lu
- Division of Medical Genetics, Pediatric Endocrinology & Metabolism, China Medical University Children’s Hospital, China Medical University, Taichung, Taiwan
| | - Hsieh-Hsun Ho
- Research and Development Department, Bioflag Biotech Co., Ltd., Tainan, Taiwan
| | - Wen-Yang Lin
- Research and Development Department, Bioflag Biotech Co., Ltd., Tainan, Taiwan
| | - Yi-Wei Kuo
- Research and Development Department, Bioflag Biotech Co., Ltd., Tainan, Taiwan
| | - Yen-Yu Huang
- Research and Development Department, Bioflag Biotech Co., Ltd., Tainan, Taiwan
| | - Shin-Yu Tsai
- Research and Development Department, Bioflag Biotech Co., Ltd., Tainan, Taiwan
| | - Hung-Chih Lin
- Division of Neonatology, China Medical University Children’s Hospital, Taichung, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
- Asia University Hospital, Asia University, Taichung, Taiwan
- *Correspondence: Hung-Chih Lin, ; orcid.org/0000-0002-8111-8371
| |
Collapse
|
31
|
Abstract
Several products consist of probiotics that are available in markets, and their potential uses are growing day by day, mainly because some strains of probiotics promote the health of gut microbiota, especially Furmicutes and Bacteroidetes, and may prevent certain gastrointestinal tract (GIT) problems. Some common diseases are inversely linked with the consumption of probiotics, i.e., obesity, type 2 diabetes, autism, osteoporosis, and some immunological disorders, for which the disease progression gets delayed. In addition to disease mitigating properties, these microbes also improve oral, nutritional, and intestinal health, followed by a robust defensive mechanism against particular gut pathogens, specifically by antimicrobial substances and peptides producing probiotics (AMPs). All these positive attributes of probiotics depend upon the type of microbial strains dispensed. Lactic acid bacteria (LAB) and Bifidobacteria are the most common microbes used, but many other microbes are available, and their use depends upon origin and health-promoting properties. This review article focuses on the most common probiotics, their health benefits, and the alleviating mechanisms against chronic kidney diseases (CKD), type 1 diabetes (T1D), type 2 diabetes (T2D), gestational diabetes mellitus (GDM), and obesity.
Collapse
|
32
|
Taylor HB, Vasu C. Impact of Prebiotic β-glucan Treatment at Juvenile Age on the Gut Microbiota Composition and the Eventual Type 1 Diabetes Onset in Non-obese Diabetic Mice. Front Nutr 2021; 8:769341. [PMID: 34805251 PMCID: PMC8595985 DOI: 10.3389/fnut.2021.769341] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022] Open
Abstract
Complex dietary polysaccharides such as β-glucans are widely used for their anti-inflammatory properties. We reported before that oral administration of Yeast β-glucan (YBG) in adult mice can help delay type 1 diabetes (T1D) onset and suppress gut inflammation through modulation of the structure and function of gut microbiota. Since juvenile age is characterized by profoundly changing immature gut microbiota, we examined the impact of oral treatment with YBG in non-obese diabetic (NOD) mice at this age. Juvenile mice that received daily oral administration of YBG starting at 15 days of age for 7 or 30 days were examined for changes in gut microbiota, immune characteristics, and T1D incidence. Mice that received YBG for 30 days but not 7 days, showed considerable changes in the composition and diversity of fecal microbiota as compared to controls. Predictive functional analysis, based on 16S rDNA sequences, revealed overrepresentation of glycan biosynthesis and metabolism, energy metabolism, and fatty acid biosynthesis pathways in mice that received YBG for 30 days. Immune phenotype of the colon showed skewing toward immune regulatory and Th17 cytokines with increases in IL-10, IL-17, and IL-21 and a decrease in TNF-α, although increases in some pro-inflammatory cytokines (IL-1b, IFN-γ) were observed. Most importantly, mice that received YBG treatment for 30 days showed significantly suppressed insulitis and delayed onset of hyperglycemia compared to controls. Overall, this study suggests that oral consumption of YBG beginning at pre-diabetic juvenile ages could have positive maturational changes to gut microbiota and immune functions and could result in a delay in the disease onset in those who are pre-disposed to T1D.
Collapse
Affiliation(s)
| | - Chenthamarakshan Vasu
- Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
33
|
Quinn LM, Wong FS, Narendran P. Environmental Determinants of Type 1 Diabetes: From Association to Proving Causality. Front Immunol 2021; 12:737964. [PMID: 34659229 PMCID: PMC8518604 DOI: 10.3389/fimmu.2021.737964] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/08/2021] [Indexed: 12/16/2022] Open
Abstract
The rising incidence of type 1 diabetes (T1D) cannot be ascribed to genetics alone, and causative environmental triggers and drivers must also be contributing. The prospective TEDDY study has provided the greatest contributions in modern time, by addressing misconceptions and refining the search strategy for the future. This review outlines the evidence to date to support the pathways from association to causality, across all stages of T1D (seroconversion to beta cell failure). We focus on infections and vaccinations; infant growth and childhood obesity; the gut microbiome and the lifestyle factors which cultivate it. Of these, the environmental determinants which have the most supporting evidence are enterovirus infection, rapid weight gain in early life, and the microbiome. We provide an infographic illustrating the key environmental determinants in T1D and their likelihood of effect. The next steps are to investigate these environmental triggers, ideally though gold-standard randomised controlled trials and further prospective studies, to help explore public health prevention strategies.
Collapse
Affiliation(s)
- Lauren M Quinn
- Institute of Immunology and Immunotherapy, Research College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom.,Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - F Susan Wong
- Department of Diabetes, University Hospitals of Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Parth Narendran
- Institute of Immunology and Immunotherapy, Research College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom.,Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
34
|
Rubin D, Bosy-Westphal A, Kabisch S, Kronsbein P, Simon MC, Tombek A, Weber K, Skurk T. Empfehlungen zur Ernährung von Personen mit Typ-1-Diabetes mellitus. DIABETOL STOFFWECHS 2021. [DOI: 10.1055/a-1515-8766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Diana Rubin
- Vivantes Klinikum Spandau, Berlin
- Vivantes Humboldt Klinikum, Berlin
| | - Anja Bosy-Westphal
- Institut für Humanernährung, Agrar- und Ernährungswissenschaftliche Fakultät, Christian-Albrechts-Universität zu Kiel, Kiel
| | - Stefan Kabisch
- Deutsches Zentrum für Diabetesforschung (DZD), München
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin, Technische Universität München, Freising
| | - Peter Kronsbein
- Fachbereich Oecotrophologie, Hochschule Niederrhein, Campus Mönchengladbach
| | - Marie-Christine Simon
- Institut für Ernährungs- und Lebensmittelwissenschaften, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn
| | | | - Katharina Weber
- Institut für Epidemiologie, Christian-Albrechts-Universität zu Kiel, Kiel
| | - Thomas Skurk
- ZIEL – Institute for Food & Health, Technische Universität München, München
- Else Kröner-Fresenius-Zentrum für Ernährungsmedizin, Technische Universität München, Freising
| | | |
Collapse
|
35
|
Alagiakrishnan K, Halverson T. Holistic perspective of the role of gut microbes in diabetes mellitus and its management. World J Diabetes 2021; 12:1463-1478. [PMID: 34630900 PMCID: PMC8472496 DOI: 10.4239/wjd.v12.i9.1463] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/24/2021] [Accepted: 08/13/2021] [Indexed: 02/06/2023] Open
Abstract
The gut microbiota (GM) plays a role in the development and progression of type 1 and type 2 diabetes mellitus (DM) and its complications. Gut dysbiosis contributes to the pathogenesis of DM. The GM has been shown to influence the efficacy of different antidiabetic medications. Intake of gut biotics, like prebiotics, probiotics and synbiotics, can improve the glucose control as well as the metabolic profile associated with DM. There is some preliminary evidence that it might even help with the cardiovascular, ophthalmic, nervous, and renal complications of DM and even contribute to the prevention of DM. More large-scale research studies are needed before wide spread use of gut biotics in clinical practice as an adjuvant therapy to the current management of DM.
Collapse
Affiliation(s)
| | - Tyler Halverson
- Department of Medicine, University of Alberta, Edmonton T6G 2G3, Alberta, Canada
| |
Collapse
|
36
|
Wu H, Chiou J. Potential Benefits of Probiotics and Prebiotics for Coronary Heart Disease and Stroke. Nutrients 2021; 13:2878. [PMID: 34445037 PMCID: PMC8401746 DOI: 10.3390/nu13082878] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 12/16/2022] Open
Abstract
Among cardiovascular diseases (CVDs), a major cause of morbidity and mortality worldwide, coronary heart disease and stroke are the most well-known and extensively studied. The onset and progression of CVD is associated with multiple risk factors, among which, gut microbiota has received much attention in the past two decades. Gut microbiota, the microbial community colonizing in the gut, plays a prominent role in human health. In particular, gut dysbiosis is directly related to many acute or chronic dysfunctions of the cardiovascular system (CVS) in the host. Earlier studies have demonstrated that the pathogenesis of CVD is strongly linked to intestinal microbiota imbalance and inflammatory responses. Probiotics and prebiotics conferring various health benefits on the host are emerging as promising therapeutic interventions for many diseases. These two types of food supplements have the potential to alleviate the risks of CVD through improving the levels of several cardiovascular markers, such as total and low-density lipoprotein (LDL) cholesterol, high sensitivity C-reactive protein (hs-CRP), and certain cytokines involved in the inflammatory response. In this review, we focus mainly on the preventive effects of probiotics and prebiotics on CVD via rebalancing the structural and functional changes in gut microbiota and maintaining immune homeostasis.
Collapse
Affiliation(s)
- Haicui Wu
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China;
- Research Institute for Future Food, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Jiachi Chiou
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China;
- Research Institute for Future Food, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| |
Collapse
|
37
|
Gut microbiota in pancreatic diseases: possible new therapeutic strategies. Acta Pharmacol Sin 2021; 42:1027-1039. [PMID: 33093569 DOI: 10.1038/s41401-020-00532-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/08/2020] [Indexed: 12/13/2022] Open
Abstract
Pancreatic diseases such as pancreatitis, type 1 diabetes and pancreatic cancer impose substantial health-care costs and contribute to marked morbidity and mortality. Recent studies have suggested a link between gut microbiota dysbiosis and pancreatic diseases; however, the potential roles and mechanisms of action of gut microbiota in pancreatic diseases remain to be fully elucidated. In this review, we summarize the evidence that supports relationship between alterations of gut microbiota and development of pancreatic diseases, and discuss the potential molecular mechanisms of gut microbiota dysbiosis in the pathogenesis of pancreatic diseases. We also propose current strategies toward gut microbiota to advance a developing research field that has clinical potential to reduce the cost of pancreatic diseases.
Collapse
|
38
|
Stojanović I, Saksida T, Miljković Đ, Pejnović N. Modulation of Intestinal ILC3 for the Treatment of Type 1 Diabetes. Front Immunol 2021; 12:653560. [PMID: 34149694 PMCID: PMC8209467 DOI: 10.3389/fimmu.2021.653560] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/18/2021] [Indexed: 11/22/2022] Open
Abstract
Gut-associated lymphoid tissue (GALT) is crucial for the maintenance of the intestinal homeostasis, but it is also the potential site of the activation of autoreactive cells and initiation/propagation of autoimmune diseases in the gut and in the distant organs. Type 3 innate lymphoid cells (ILC3) residing in the GALT integrate signals from food ingredients and gut microbiota metabolites in order to control local immunoreactivity. Notably, ILC3 secrete IL-17 and GM-CSF that activate immune cells in combating potentially pathogenic microorganisms. ILC3 also produce IL-22 that potentiates the strength and integrity of epithelial tight junctions, production of mucus and antimicrobial peptides thus enabling the proper function of the intestinal barrier. The newly discovered function of small intestine ILC3 is the secretion of IL-2 and the promotion of regulatory T cell (Treg) generation and function. Since the intestinal barrier dysfunction, together with the reduction in small intestine ILC3 and Treg numbers are associated with the pathogenesis of type 1 diabetes (T1D), the focus of this article is intestinal ILC3 modulation for the therapy of T1D. Of particular interest is free fatty acids receptor 2 (FFAR2), predominantly expressed on intestinal ILC3, that can be stimulated by available selective synthetic agonists. Thus, we propose that FFAR2-based interventions by boosting ILC3 beneficial functions may attenuate autoimmune response against pancreatic β cells during T1D. Also, it is our opinion that treatments based on ILC3 stimulation by functional foods can be used as prophylaxis in individuals that are genetically predisposed to develop T1D.
Collapse
Affiliation(s)
- Ivana Stojanović
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Tamara Saksida
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Đorđe Miljković
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Nada Pejnović
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
39
|
FFAR from the Gut Microbiome Crowd: SCFA Receptors in T1D Pathology. Metabolites 2021; 11:metabo11050302. [PMID: 34064625 PMCID: PMC8151283 DOI: 10.3390/metabo11050302] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 12/14/2022] Open
Abstract
The gut microbiome has emerged as a novel determinant of type 1 diabetes (T1D), but the underlying mechanisms are unknown. In this context, major gut microbial metabolites, short-chain fatty acids (SCFAs), are considered to be an important link between the host and gut microbiome. We, along with other laboratories, have explored how SCFAs and their cognate receptors affect various metabolic conditions, including obesity, type 2 diabetes, and metabolic syndrome. Though gut microbiome and SCFA-level changes have been reported in T1D and in mouse models of the disease, the role of SCFA receptors in T1D remains under explored. In this review article, we will highlight the existing and possible roles of these receptors in T1D pathology. We conclude with a discussion of SCFA receptors as therapeutic targets for T1D, exploring an exciting new potential for novel treatments of glucometabolic disorders.
Collapse
|
40
|
Yoha KS, Nida S, Dutta S, Moses JA, Anandharamakrishnan C. Targeted Delivery of Probiotics: Perspectives on Research and Commercialization. Probiotics Antimicrob Proteins 2021; 14:15-48. [PMID: 33904011 PMCID: PMC8075719 DOI: 10.1007/s12602-021-09791-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2021] [Indexed: 02/07/2023]
Abstract
Considering the significance of the gut microbiota on human health, there has been ever-growing research and commercial interest in various aspects of probiotic functional foods and drugs. A probiotic food requires cautious consideration in terms of strain selection, appropriate process and storage conditions, cell viability and functionality, and effective delivery at the targeted site. To address these challenges, several technologies have been explored and some of them have been adopted for industrial applicability. Encapsulation of probiotics has been recognized as an effective way to stabilize them in their dried form. By conferring a physical barrier to protect them from adverse conditions, the encapsulation approach renders direct benefits on stability, delivery, and functionality. Various techniques have been explored to encapsulate probiotics, but it is noteworthy that the encapsulation method itself influences surface morphology, viability, and survivability of probiotics. This review focuses on the need to encapsulate probiotics, trends in various encapsulation techniques, current research and challenges in targeted delivery, the market status of encapsulated probiotics, and future directions. Specific focus has been given on various in vitro methods that have been explored to better understand their delivery and performance.
Collapse
Affiliation(s)
- K S Yoha
- Computational Modeling and Nanoscale Processing Unit, Indian Institute of Food Processing Technology (IIFPT), Ministry of Food Processing Industries, Government of India, 613 005, Thanjavur, Tamil Nadu, India
| | - Sundus Nida
- Computational Modeling and Nanoscale Processing Unit, Indian Institute of Food Processing Technology (IIFPT), Ministry of Food Processing Industries, Government of India, 613 005, Thanjavur, Tamil Nadu, India
| | - Sayantani Dutta
- Computational Modeling and Nanoscale Processing Unit, Indian Institute of Food Processing Technology (IIFPT), Ministry of Food Processing Industries, Government of India, 613 005, Thanjavur, Tamil Nadu, India
| | - J A Moses
- Computational Modeling and Nanoscale Processing Unit, Indian Institute of Food Processing Technology (IIFPT), Ministry of Food Processing Industries, Government of India, 613 005, Thanjavur, Tamil Nadu, India
| | - C Anandharamakrishnan
- Computational Modeling and Nanoscale Processing Unit, Indian Institute of Food Processing Technology (IIFPT), Ministry of Food Processing Industries, Government of India, 613 005, Thanjavur, Tamil Nadu, India.
| |
Collapse
|
41
|
Djekkoun N, Lalau JD, Bach V, Depeint F, Khorsi-Cauet H. Chronic oral exposure to pesticides and their consequences on metabolic regulation: role of the microbiota. Eur J Nutr 2021; 60:4131-4149. [PMID: 33837455 DOI: 10.1007/s00394-021-02548-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/22/2021] [Indexed: 12/17/2022]
Abstract
Pesticides have long been used in agriculture and household treatments. Pesticide residues can be found in biological samples for both the agriculture workers through direct exposure but also to the general population by indirect exposure. There is also evidence of pesticide contamination in utero and trans-generational impacts. Whilst acute exposure to pesticides has long been associated with endocrine perturbations, chronic exposure with low doses also increases the prevalence of metabolic disorders such as obesity or type 2 diabetes. Dysmetabolism is a low-grade inflammation disorder and as such the microbiota plays a role in its etiology. It is therefore important to fully understand the role of microbiota on the genesis of subsequent health effects. The digestive tract and mostly microbiota are the first organs of contact after oral exposure. The objective of this review is thus to better understand mechanisms that link pesticide exposure, dysmetabolism and microbiota. One of the key outcomes on the microbiota is the reduced Bacteroidetes and increased Firmicutes phyla, reflecting both pesticide exposure and risk factors of dysmetabolism. Other bacterial genders and metabolic activities are also involved. As for most pathologies impacting microbiota (including inflammatory disorders), the role of prebiotics can be suggested as a prevention strategy and some preliminary evidence reinforces this axis.
Collapse
Affiliation(s)
- Narimane Djekkoun
- PeriTox UMR_I 01 Laboratory, University Center for Health Research, CURS-UPJV, Picardy Jules Verne University, 80054, Amiens cedex 1, France
| | - Jean-Daniel Lalau
- PeriTox UMR_I 01 Laboratory, University Center for Health Research, CURS-UPJV, Picardy Jules Verne University, 80054, Amiens cedex 1, France.,Service Endocrinologie, Diabétologie, Nutrition, CHU Amiens Picardie, Site Nord, 80054, Amiens cedex 1, France
| | - Véronique Bach
- PeriTox UMR_I 01 Laboratory, University Center for Health Research, CURS-UPJV, Picardy Jules Verne University, 80054, Amiens cedex 1, France
| | - Flore Depeint
- Unité Transformations & Agroressources ULR7519, Institut Polytechnique UniLaSalle-Université d'Artois, 60026, Beauvais, France
| | - Hafida Khorsi-Cauet
- PeriTox UMR_I 01 Laboratory, University Center for Health Research, CURS-UPJV, Picardy Jules Verne University, 80054, Amiens cedex 1, France.
| |
Collapse
|
42
|
Gradisteanu Pircalabioru G, Corcionivoschi N, Gundogdu O, Chifiriuc MC, Marutescu LG, Ispas B, Savu O. Dysbiosis in the Development of Type I Diabetes and Associated Complications: From Mechanisms to Targeted Gut Microbes Manipulation Therapies. Int J Mol Sci 2021; 22:2763. [PMID: 33803255 PMCID: PMC7967220 DOI: 10.3390/ijms22052763] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/06/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Globally, we are facing a worrying increase in type 1 diabetes mellitus (T1DM) incidence, with onset at younger age shedding light on the need to better understand the mechanisms of disease and step-up prevention. Given its implication in immune system development and regulation of metabolism, there is no surprise that the gut microbiota is a possible culprit behind T1DM pathogenesis. Additionally, microbiota manipulation by probiotics, prebiotics, dietary factors and microbiota transplantation can all modulate early host-microbiota interactions by enabling beneficial microbes with protective potential for individuals with T1DM or at high risk of developing T1DM. In this review, we discuss the challenges and perspectives of translating microbiome data into clinical practice. Nevertheless, this progress will only be possible if we focus our interest on developing numerous longitudinal, multicenter, interventional and double-blind randomized clinical trials to confirm their efficacy and safety of these therapeutic approaches.
Collapse
Affiliation(s)
| | - Nicolae Corcionivoschi
- Bacteriology Branch, Veterinary Sciences Division, Agri-Food and Biosciences Institute, Belfast BT9 5PX, UK;
| | - Ozan Gundogdu
- Faculty of Infectious & Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK;
| | - Mariana-Carmen Chifiriuc
- Research Institute of University of Bucharest (ICUB), 300645 Bucharest, Romania; (G.G.P.); (L.G.M.); (B.I.)
- Academy of Romanian Scientists, 050045 Bucharest, Romania
| | | | - Bogdan Ispas
- Research Institute of University of Bucharest (ICUB), 300645 Bucharest, Romania; (G.G.P.); (L.G.M.); (B.I.)
| | - Octavian Savu
- “N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 2nd District, 020042 Bucharest, Romania;
- Department of Doctoral School, “Carol Davila” University of Medicine and Pharmacy, 5th District, 050474 Bucharest, Romania
| |
Collapse
|
43
|
Kataoka K, Nemoto H, Sakurai A, Yasutomo K, Shikanai M. Preventive effect of fermented brown rice and rice bran on spontaneous type 1 diabetes in NOD female mice. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
44
|
Groele L, Szajewska H, Szalecki M, Świderska J, Wysocka-Mincewicz M, Ochocińska A, Stelmaszczyk-Emmel A, Demkow U, Szypowska A. Lack of effect of Lactobacillus rhamnosus GG and Bifidobacterium lactis Bb12 on beta-cell function in children with newly diagnosed type 1 diabetes: a randomised controlled trial. BMJ Open Diabetes Res Care 2021; 9:9/1/e001523. [PMID: 33771763 PMCID: PMC8006832 DOI: 10.1136/bmjdrc-2020-001523] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 02/25/2021] [Accepted: 02/28/2021] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION The gut microbiota may be relevant in the development of type 1 diabetes (T1D). We examined the effects of Lactobacillus rhamnosus GG and Bifidobacterium lactis Bb12 on beta-cell function in children with newly diagnosed T1D. RESEARCH DESIGN AND METHODS Children aged 8-17 years with newly (within 60 days) diagnosed T1D were enrolled in a double-blind, randomised controlled trial in which they received L. rhamnosus GG and B. lactis Bb12 at a dose of 109 colony-forming units or placebo, orally, once daily, for 6 months. The follow-up was for 12 months. The primary outcome measure was the area under the curve (AUC) of the C-peptide level during 2-hour responses to a mixed meal. RESULTS Ninety-six children were randomised (probiotics, n=48; placebo n=48; median age 12.3 years). Eighty-eight (92%) completed the 6-month intervention, and 87 (91%) completed the follow-up at 12 months. There was no significant difference between the study groups for the AUC of the C-peptide level. For the secondary outcomes at 6 months, there were no differences between the study groups. At 12 months, with one exception, there also were no significant differences between the groups. Compared with the placebo group, there was a significantly increased number of subjects with thyroid autoimmunity in the probiotic group. However, at baseline, there was also a higher frequency of thyroid autoimmunity in the probiotic group. There were no cases of severe hypoglycemia or ketoacidosis in any of the groups. No adverse events related to the study products were reported. CONCLUSIONS L. rhamnosus GG and B. lactis Bb12, as administered in this study, had no significant effect in maintaining the residual pancreatic beta-cell function in children with newly diagnosed T1D. It remains unclear which probiotics, if any, alone or in combination, are potentially the most useful for management of T1D. TRIAL REGISTRATION NUMBER NCT03032354.
Collapse
Affiliation(s)
- Lidia Groele
- Department of Paediatrics, The Children's Clinical Hospital Józef Polikarp Brudziński, Warsaw, Poland
| | - Hania Szajewska
- Department of Paediatrics, The Medical University of Warsaw, Warsaw, Poland
| | - Mieczysław Szalecki
- Clinic of Endocrinology and Diabetology, The Children's Memorial Health Institute, Warsaw, Poland
- Collegium Medicum (Jan Kochanowski University), Kielce, Poland
| | - Jolanta Świderska
- Clinic of Endocrinology and Diabetology, The Children's Memorial Health Institute, Warsaw, Poland
| | - Marta Wysocka-Mincewicz
- Clinic of Endocrinology and Diabetology, The Children's Memorial Health Institute, Warsaw, Poland
| | - Agnieszka Ochocińska
- Department of Biochemistry, Radioimmunology and Experimental Medicine, The Children's Memorial Health Institute, Warsaw, Poland
| | - Anna Stelmaszczyk-Emmel
- Deparment of Laboratory Diagnostics and Clinical Immunology of Developmental Age, The Medical University of Warsaw, Warsaw, Poland
| | - Urszula Demkow
- Deparment of Laboratory Diagnostics and Clinical Immunology of Developmental Age, The Medical University of Warsaw, Warsaw, Poland
| | | |
Collapse
|
45
|
Abstract
Gut dysbiosis in diabetes mellitus is associated with decreased short-chain fatty acids and epithelial barrier disruption. Microbial-derived toxins move across the "leaky gut" and incur systemic inflammation and insulin resistance. In children, gut dysbiosis has been associated with risk of developing type 1 diabetes mellitus. In animal models, the obesity phenotype is transferable via microbiota transplantation. Plant-based low protein diets and certain anti-diabetic drugs have been associated with positive microbiome effects. Clinical trials with prebiotics and probiotics have yielded mixed results. Further investigations are needed to evaluate the gut microbiome as a potential therapeutic target for diabetes prevention and management.
Collapse
Affiliation(s)
- Wei Ling Lau
- Division of Nephrology, University of California, Irvine School of Medicine, Orange, CA.
| | - Tiffany Tran
- Division of Nephrology, University of California, Irvine School of Medicine, Orange, CA
| | - Connie M Rhee
- Harold Simmons Center for Kidney Disease Research and Epidemiology, Division of Nephrology, University of California, Irvine School of Medicine, Orange, CA
| | - Kamyar Kalantar-Zadeh
- Harold Simmons Center for Kidney Disease Research and Epidemiology, Division of Nephrology, University of California, Irvine School of Medicine, Orange, CA
| | - Nosratola D Vaziri
- Division of Nephrology, University of California, Irvine School of Medicine, Orange, CA
| |
Collapse
|
46
|
Webb‐Robertson BM, Bramer LM, Stanfill BA, Reehl SM, Nakayasu ES, Metz TO, Frohnert BI, Norris JM, Johnson RK, Rich SS, Rewers MJ. Prediction of the development of islet autoantibodies through integration of environmental, genetic, and metabolic markers. J Diabetes 2021; 13:143-153. [PMID: 33124145 PMCID: PMC7818425 DOI: 10.1111/1753-0407.13093] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/29/2020] [Accepted: 07/15/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The Environmental Determinants of the Diabetes in the Young (TEDDY) study has prospectively followed, from birth, children at increased genetic risk of type 1 diabetes. TEDDY has collected heterogenous data longitudinally to gain insights into the environmental and biological mechanisms driving the progression to persistent islet autoantibodies. METHODS We developed a machine learning model to predict imminent transition to the development of persistent islet autoantibodies based on time-varying metabolomics data integrated with time-invariant risk factors (eg, gestational age). The machine learning was initiated with 221 potential features (85 genetic, 5 environmental, 131 metabolomic) and an ensemble-based feature evaluation was utilized to identify a small set of predictive features that can be interrogated to better understand the pathogenesis leading up to persistent islet autoimmunity. RESULTS The final integrative machine learning model included 42 disparate features, returning a cross-validated receiver operating characteristic area under the curve (AUC) of 0.74 and an AUC of ~0.65 on an independent validation dataset. The model identified a principal set of 20 time-invariant markers, including 18 genetic markers (16 single nucleotide polymorphisms [SNPs] and two HLA-DR genotypes) and two demographic markers (gestational age and exposure to a prebiotic formula). Integration with the metabolome identified 22 supplemental metabolites and lipids, including adipic acid and ceramide d42:0, that predicted development of islet autoantibodies. CONCLUSIONS The majority (86%) of metabolites that predicted development of islet autoantibodies belonged to three pathways: lipid oxidation, phospholipase A2 signaling, and pentose phosphate, suggesting that these metabolic processes may play a role in triggering islet autoimmunity.
Collapse
Affiliation(s)
- Bobbie‐Jo M. Webb‐Robertson
- Biological Sciences Division, Pacific Northwest National LaboratoryRichlandWashingtonUSA
- Colorado School of Public HealthUniversity of Colorado Anschutz Medical CampusAuroraCaliforniaUSA
| | - Lisa M. Bramer
- Computing and Analytics DivisionPacific Northwest National LaboratoryRichlandWashingtonUSA
| | - Bryan A. Stanfill
- Computing and Analytics DivisionPacific Northwest National LaboratoryRichlandWashingtonUSA
| | - Sarah M. Reehl
- Computing and Analytics DivisionPacific Northwest National LaboratoryRichlandWashingtonUSA
| | - Ernesto S. Nakayasu
- Biological Sciences Division, Pacific Northwest National LaboratoryRichlandWashingtonUSA
| | - Thomas O. Metz
- Biological Sciences Division, Pacific Northwest National LaboratoryRichlandWashingtonUSA
| | - Brigitte I. Frohnert
- Barbara Davis Center for DiabetesUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Jill M. Norris
- Colorado School of Public HealthUniversity of Colorado Anschutz Medical CampusAuroraCaliforniaUSA
| | - Randi K. Johnson
- Colorado School of Public HealthUniversity of Colorado Anschutz Medical CampusAuroraCaliforniaUSA
| | - Stephen S. Rich
- Center for Public Health GenomicsUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Marian J. Rewers
- Barbara Davis Center for DiabetesUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| |
Collapse
|
47
|
Mishra SP, Jain S, Taraphder S, Yadav H. New Horizons in Microbiota and Metabolic Health Research. J Clin Endocrinol Metab 2021; 106:e1052-e1059. [PMID: 33128374 PMCID: PMC7823252 DOI: 10.1210/clinem/dgaa769] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Indexed: 01/02/2023]
Abstract
Decade-old studies have demonstrated that microbes living in our gut (microbiota) contribute to both maintaining normal metabolic function and to the pathology of metabolic diseases, such as obesity and diabetes. Emerging evidence suggests that gut microbiota influences the personalized effects of diets and drugs and impact the gut-brain axis and leaky gut inflammation to control metabolic function/diseases. Gut microbiota can be an ideal source of prognostic markers and therapies for metabolic diseases. Here we discuss the emerging concepts in the area of microbiota and metabolic interactions in personalized nutrition, drug response, and disease prognosis.
Collapse
Affiliation(s)
- Sidharth P Mishra
- Department of Internal Medicine–Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Department of Animal Genetics and Breeding, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal, India
| | - Shalini Jain
- Metabolic Phenotyping Shared Resource, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Department of Internal Medicine-Endocrinology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Subhash Taraphder
- Department of Animal Genetics and Breeding, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal, India
| | - Hariom Yadav
- Department of Internal Medicine–Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
48
|
Verduci E, Mameli C, Amatruda M, Petitti A, Vizzuso S, El Assadi F, Zuccotti G, Alabduljabbar S, Terranegra A. Early Nutrition and Risk of Type 1 Diabetes: The Role of Gut Microbiota. Front Nutr 2021; 7:612377. [PMID: 33425976 PMCID: PMC7785819 DOI: 10.3389/fnut.2020.612377] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022] Open
Abstract
Type 1 diabetes (T1D) appears most frequently in childhood, with an alarming increasing incidence in the last decades. Although the genetic predisposition is a major risk factor, it cannot solely explain the complex etiology of T1D which is still not fully understood. In this paper, we reviewed the most recent findings on the role of early nutrition and the involvement of the gut microbiota in the etiopathogenesis of T1D. The main conclusions that are withdrawn from the current literature regarding alleviating the risk of developing T1D through nutrition are the encouragement of long-term breast-feeding for at least the first 6 months of life and the avoidance of early complementary foods and gluten introduction (before 4 months of age) as well as cow milk introduction before 12 months of life. These detrimental feeding habits create a gut microbiota dysbiotic state that can contribute to the onset of T1D in infancy. Finally, we discussed the possibility to introduce probiotics, prebiotics and post-biotics in the prevention of T1D.
Collapse
Affiliation(s)
- Elvira Verduci
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy.,Department of Health Sciences, University of Milan, Milan, Italy
| | - Chiara Mameli
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy
| | - Matilde Amatruda
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Agnese Petitti
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy
| | - Sara Vizzuso
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Farah El Assadi
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Gianvincenzo Zuccotti
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy
| | | | | |
Collapse
|
49
|
The Gut Microbiota Profile According to Glycemic Control in Type 1 Diabetes Patients Treated with Personal Insulin Pumps. Microorganisms 2021; 9:microorganisms9010155. [PMID: 33445500 PMCID: PMC7826603 DOI: 10.3390/microorganisms9010155] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/23/2020] [Accepted: 01/08/2021] [Indexed: 12/12/2022] Open
Abstract
Recently, several studies explored associations between type 1 diabetes (T1DM) and microbiota. The aim of our study was to assess the colonic microbiota structure according to the metabolic control in T1DM patients treated with insulin pumps. We studied 89 T1DM patients (50.6% women) at the median age of 25 (IQR, 22-29) years. Pielou's evenness (p = 0.02), and Shannon's (p = 0.04) and Simpson's diversity indexes (p = 0.01), were higher in patients with glycosylated hemoglobin (HbA1c) ≥ 53 mmol/mol (7%). There were no differences in beta diversity between groups. A linear discriminant analysis effect size (LEfSe) algorithm showed that one family (Ruminococcaceae) was enriched in patients with HbA1c < 53 mmol/mol, whereas one family (Streptococcaceae) and four species (Ruminococcus torques, unclassified species of Lactococcus, Eubacteroim dolichum, and Coprobacillus cateniformis) were enriched in patients with HbA1c ≥ 53 mmol/mol. We found that at class level, the following pathways according to Kyoto Encyclopedia of Genes and Genomes were enriched in patients with HbA1c < 53 mmol/mol: bacterial motility proteins, secretion system, bacterial secretion system, ribosome biogenesis, translation proteins, and lipid biosynthesis, whereas in patients with HbA1c ≥ 53 mmol/mol, the galactose metabolism, oxidative phosphorylation, phosphotransferase system, fructose, and mannose metabolism were enriched. Observed differences in alpha diversity, metabolic pathways, and associations between bacteria and HbA1c in colonic flora need further investigation.
Collapse
|
50
|
Elhag DA, Kumar M, Al Khodor S. Exploring the Triple Interaction between the Host Genome, the Epigenome, and the Gut Microbiome in Type 1 Diabetes. Int J Mol Sci 2020; 22:ijms22010125. [PMID: 33374418 PMCID: PMC7795494 DOI: 10.3390/ijms22010125] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 12/13/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022] Open
Abstract
Type 1 diabetes (T1D) is an auto-immune disorder characterized by a complex interaction between the host immune system and various environmental factors in genetically susceptible individuals. Genome-wide association studies (GWAS) identified different T1D risk and protection alleles, however, little is known about the environmental factors that can be linked to these alleles. Recent evidence indicated that, among those environmental factors, dysbiosis (imbalance) in the gut microbiota may play a role in the pathogenesis of T1D, affecting the integrity of the gut and leading to systemic inflammation and auto-destruction of the pancreatic β cells. Several studies have identified changes in the gut microbiome composition in humans and animal models comparing T1D subjects with controls. Those changes were characterized by a higher abundance of Bacteroides and a lower abundance of the butyrate-producing bacteria such as Clostridium clusters IV and XIVa. The mechanisms by which the dysbiotic bacteria and/or their metabolites interact with the genome and/or the epigenome of the host leading to destructive autoimmunity is still not clear. As T1D is a multifactorial disease, understanding the interaction between different environmental factors such as the gut microbiome, the genetic and the epigenetic determinants that are linked with the early appearance of autoantibodies can expand our knowledge about the disease pathogenesis. This review aims to provide insights into the interaction between the gut microbiome, susceptibility genes, epigenetic factors, and the immune system in the pathogenesis of T1D.
Collapse
|