1
|
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
|
2
|
Pant T, Lin CW, Bedrat A, Jia S, Roethle MF, Truchan NA, Ciecko AE, Chen YG, Hessner MJ. Monocytes in type 1 diabetes families exhibit high cytolytic activity and subset abundances that correlate with clinical progression. SCIENCE ADVANCES 2024; 10:eadn2136. [PMID: 38758799 PMCID: PMC11100571 DOI: 10.1126/sciadv.adn2136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 04/15/2024] [Indexed: 05/19/2024]
Abstract
Monocytes are immune regulators implicated in the pathogenesis of type 1 diabetes (T1D), an autoimmune disease that targets insulin-producing pancreatic β cells. We determined that monocytes of recent onset (RO) T1D patients and their healthy siblings express proinflammatory/cytolytic transcriptomes and hypersecrete cytokines in response to lipopolysaccharide exposure compared to unrelated healthy controls (uHCs). Flow cytometry measured elevated circulating abundances of intermediate monocytes and >2-fold more CD14+CD16+HLADR+KLRD1+PRF1+ NK-like monocytes among patients with ROT1D compared to uHC. The intermediate to nonclassical monocyte ratio among ROT1D patients correlated with the decline in functional β cell mass during the first 24 months after onset. Among sibling nonprogressors, temporal decreases were measured in the intermediate to nonclassical monocyte ratio and NK-like monocyte abundances; these changes coincided with increases in activated regulatory T cells. In contrast, these monocyte populations exhibited stability among T1D progressors. This study associates heightened monocyte proinflammatory/cytolytic activity with T1D susceptibility and progression and offers insight to the age-dependent decline in T1D susceptibility.
Collapse
Affiliation(s)
- Tarun Pant
- The Max McGee Research Center for Juvenile Diabetes, Children’s Research Institute of Children’s Hospital of Wisconsin, Milwaukee, WI, USA
- Department of Pediatrics, The Medical College of Wisconsin, Milwaukee, WI, USA
| | - Chien-Wei Lin
- Division of Biostatistics, The Medical College of Wisconsin, Milwaukee, WI, USA
| | - Amina Bedrat
- The Max McGee Research Center for Juvenile Diabetes, Children’s Research Institute of Children’s Hospital of Wisconsin, Milwaukee, WI, USA
- Department of Pediatrics, The Medical College of Wisconsin, Milwaukee, WI, USA
| | - Shuang Jia
- The Max McGee Research Center for Juvenile Diabetes, Children’s Research Institute of Children’s Hospital of Wisconsin, Milwaukee, WI, USA
- Department of Pediatrics, The Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mark F. Roethle
- The Max McGee Research Center for Juvenile Diabetes, Children’s Research Institute of Children’s Hospital of Wisconsin, Milwaukee, WI, USA
- Department of Pediatrics, The Medical College of Wisconsin, Milwaukee, WI, USA
| | - Nathan A. Truchan
- The Max McGee Research Center for Juvenile Diabetes, Children’s Research Institute of Children’s Hospital of Wisconsin, Milwaukee, WI, USA
- Department of Pediatrics, The Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ashley E. Ciecko
- The Max McGee Research Center for Juvenile Diabetes, Children’s Research Institute of Children’s Hospital of Wisconsin, Milwaukee, WI, USA
- Department of Pediatrics, The Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yi-Guang Chen
- The Max McGee Research Center for Juvenile Diabetes, Children’s Research Institute of Children’s Hospital of Wisconsin, Milwaukee, WI, USA
- Department of Pediatrics, The Medical College of Wisconsin, Milwaukee, WI, USA
| | - Martin J. Hessner
- The Max McGee Research Center for Juvenile Diabetes, Children’s Research Institute of Children’s Hospital of Wisconsin, Milwaukee, WI, USA
- Department of Pediatrics, The Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
3
|
Pöysti S, Silojärvi S, Brodnicki TC, Catterall T, Liu X, Mackin L, Luster AD, Kay TWH, Christen U, Thomas HE, Hänninen A. Gut dysbiosis promotes islet-autoimmunity by increasing T-cell attraction in islets via CXCL10 chemokine. J Autoimmun 2023; 140:103090. [PMID: 37572540 DOI: 10.1016/j.jaut.2023.103090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/29/2023] [Accepted: 07/11/2023] [Indexed: 08/14/2023]
Abstract
CXCL10 is an IFNγ-inducible chemokine implicated in the pathogenesis of type 1 diabetes. T-cells attracted to pancreatic islets produce IFNγ, but it is unclear what attracts the first IFNγ -producing T-cells in islets. Gut dysbiosis following administration of pathobionts induced CXCL10 expression in pancreatic islets of healthy non-diabetes-prone (C57BL/6) mice and depended on TLR4-signaling, and in non-obese diabetic (NOD) mice, gut dysbiosis induced also CXCR3 chemokine receptor in IGRP-reactive islet-specific T-cells in pancreatic lymph node. In amounts typical to low-grade endotoxemia, bacterial lipopolysaccharide induced CXCL10 production in isolated islets of wild type and RAG1 or IFNG-receptor-deficient but not type-I-IFN-receptor-deficient NOD mice, dissociating lipopolysaccharide-induced CXCL10 production from T-cells and IFNγ. Although mostly myeloid-cell dependent, also β-cells showed activation of innate immune signaling pathways and Cxcl10 expression in response to lipopolysaccharide indicating their independent sensitivity to dysbiosis. Thus, CXCL10 induction in response to low levels of lipopolysaccharide may allow islet-specific T-cells imprinted in pancreatic lymph node to enter in healthy islets independently of IFN-g, and thus link gut dysbiosis to early islet-autoimmunity via dysbiosis-associated low-grade endotoxemia.
Collapse
MESH Headings
- Animals
- Mice
- Autoimmunity
- Chemokine CXCL10/metabolism
- Chemokine CXCL10/immunology
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/etiology
- Disease Models, Animal
- Dysbiosis/immunology
- Gastrointestinal Microbiome/immunology
- Interferon-gamma/metabolism
- Islets of Langerhans/immunology
- Islets of Langerhans/metabolism
- Lipopolysaccharides/immunology
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Knockout
- Receptors, CXCR3/metabolism
- Receptors, CXCR3/genetics
- Receptors, CXCR3/immunology
- Signal Transduction
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Toll-Like Receptor 4/metabolism
Collapse
Affiliation(s)
- Sakari Pöysti
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Satu Silojärvi
- Institute of Biomedicine, University of Turku, Turku, Finland
| | | | - Tara Catterall
- St. Vincent's Institute of Medical Research, Melbourne, VIC, Australia
| | - Xin Liu
- St. Vincent's Institute of Medical Research, Melbourne, VIC, Australia
| | - Leanne Mackin
- St. Vincent's Institute of Medical Research, Melbourne, VIC, Australia
| | - Andrew D Luster
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Thomas W H Kay
- St. Vincent's Institute of Medical Research, Melbourne, VIC, Australia
| | - Urs Christen
- Klinikum der Goethe Universität Frankfurt, Frankfurt Am Main, Germany
| | - Helen E Thomas
- St. Vincent's Institute of Medical Research, Melbourne, VIC, Australia
| | - Arno Hänninen
- Institute of Biomedicine, University of Turku, Turku, Finland; Turku University Hospital Laboratory Division, Turku, Finland.
| |
Collapse
|
4
|
Buckner T, Johnson RK, Vanderlinden LA, Carry PM, Romero A, Onengut-Gumuscu S, Chen WM, Fiehn O, Frohnert BI, Crume T, Perng W, Kechris K, Rewers M, Norris JM. An Oxylipin-Related Nutrient Pattern and Risk of Type 1 Diabetes in the Diabetes Autoimmunity Study in the Young (DAISY). Nutrients 2023; 15:945. [PMID: 36839302 PMCID: PMC9962656 DOI: 10.3390/nu15040945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Oxylipins, pro-inflammatory and pro-resolving lipid mediators, are associated with the risk of type 1 diabetes (T1D) and may be influenced by diet. This study aimed to develop a nutrient pattern related to oxylipin profiles and test their associations with the risk of T1D among youth. The nutrient patterns were developed with a reduced rank regression in a nested case-control study (n = 335) within the Diabetes Autoimmunity Study in the Young (DAISY), a longitudinal cohort of children at risk of T1D. The oxylipin profiles (adjusted for genetic predictors) were the response variables. The nutrient patterns were tested in the case-control study (n = 69 T1D cases, 69 controls), then validated in the DAISY cohort using a joint Cox proportional hazards model (n = 1933, including 81 T1D cases). The first nutrient pattern (NP1) was characterized by low beta cryptoxanthin, flavanone, vitamin C, total sugars and iron, and high lycopene, anthocyanidins, linoleic acid and sodium. After adjusting for T1D family history, the HLA genotype, sex and race/ethnicity, NP1 was associated with a lower risk of T1D in the nested case-control study (OR: 0.44, p = 0.0126). NP1 was not associated with the risk of T1D (HR: 0.54, p-value = 0.1829) in the full DAISY cohort. Future studies are needed to confirm the nested case-control findings and investigate the modifiable factors for oxylipins.
Collapse
Affiliation(s)
- Teresa Buckner
- Department of Epidemiology, Colorado School of Public Health, CU Anschutz, Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Kinesiology, Nutrition, and Dietetics, University of Northern Colorado, Greeley, CO 80639, USA
| | - Randi K. Johnson
- Department of Epidemiology, Colorado School of Public Health, CU Anschutz, Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Biomedical Informatics, CU School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Lauren A. Vanderlinden
- Department of Epidemiology, Colorado School of Public Health, CU Anschutz, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Patrick M. Carry
- Department of Epidemiology, Colorado School of Public Health, CU Anschutz, Anschutz Medical Campus, Aurora, CO 80045, USA
- Colorado Program for Musculoskeletal Research, Department of Orthopedics, CU School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Alex Romero
- Department of Biomedical Informatics, CU School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Suna Onengut-Gumuscu
- Health Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22903, USA
| | - Wei-Min Chen
- Health Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22903, USA
| | - Oliver Fiehn
- NIH-West Coast Metabolomics Center, University of California-Davis, Davis, CA 95616, USA
| | - Brigitte I. Frohnert
- Department of Epidemiology, Colorado School of Public Health, CU Anschutz, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Tessa Crume
- Department of Epidemiology, Colorado School of Public Health, CU Anschutz, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Wei Perng
- Department of Epidemiology, Colorado School of Public Health, CU Anschutz, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Katerina Kechris
- Department of Epidemiology, Colorado School of Public Health, CU Anschutz, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Marian Rewers
- Department of Epidemiology, Colorado School of Public Health, CU Anschutz, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jill M. Norris
- Department of Epidemiology, Colorado School of Public Health, CU Anschutz, Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
5
|
Pritchard KA, Jing X, Teng M, Wells C, Jia S, Afolayan AJ, Jarzembowski J, Day BW, Naylor S, Hessner MJ, Konduri GG, Teng RJ. Role of endoplasmic reticulum stress in impaired neonatal lung growth and bronchopulmonary dysplasia. PLoS One 2022; 17:e0269564. [PMID: 36018859 PMCID: PMC9417039 DOI: 10.1371/journal.pone.0269564] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/24/2022] [Indexed: 11/18/2022] Open
Abstract
Myeloperoxidase (MPO), oxidative stress (OS), and endoplasmic reticulum (ER) stress are increased in the lungs of rat pups raised in hyperoxia, an established model of bronchopulmonary dysplasia (BPD). However, the relationship between OS, MPO, and ER stress has not been examined in hyperoxia rat pups. We treated Sprague-Dawley rat pups with tunicamycin or hyperoxia to determine this relationship. ER stress was detected using immunofluorescence, transcriptomic, proteomic, and electron microscopic analyses. Immunofluorescence observed increased ER stress in the lungs of hyperoxic rat BPD and human BPD. Proteomic and morphometric studies showed that tunicamycin directly increased ER stress of rat lungs and decreased lung complexity with a BPD phenotype. Previously, we showed that hyperoxia initiates a cycle of destruction that we hypothesized starts from increasing OS through MPO accumulation and then increases ER stress to cause BPD. To inhibit ER stress, we used tauroursodeoxycholic acid (TUDCA), a molecular chaperone. To break the cycle of destruction and reduce OS and MPO, we used N-acetyl-lysyltyrosylcysteine amide (KYC). The fact that TUDCA improved lung complexity in tunicamycin- and hyperoxia-treated rat pups supports the idea that ER stress plays a causal role in BPD. Additional support comes from data showing TUDCA decreased lung myeloid cells and MPO levels in the lungs of tunicamycin- and hyperoxia-treated rat pups. These data link OS and MPO to ER stress in the mechanisms mediating BPD. KYC's inhibition of ER stress in the tunicamycin-treated rat pup's lung provides additional support for the idea that MPO-induced ER stress plays a causal role in the BPD phenotype. ER stress appears to expand our proposed cycle of destruction. Our results suggest ER stress evolves from OS and MPO to increase neonatal lung injury and impair growth and development. The encouraging effect of TUDCA indicates that this compound has the potential for treating BPD.
Collapse
Affiliation(s)
- Kirkwood A. Pritchard
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America,Children’s Research Institute, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
| | - Xigang Jing
- Children’s Research Institute, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America,Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
| | - Michelle Teng
- Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
| | - Clive Wells
- Electron Microscope Facility, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
| | - Shuang Jia
- Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
| | - Adeleye J. Afolayan
- Children’s Research Institute, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America,Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
| | - Jason Jarzembowski
- Children’s Research Institute, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America,Division of Pediatric Pathology, Department of Pathology, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
| | - Billy W. Day
- ReNeuroGen L.L.C. Milwaukee, Elm Grove, Wisconsin, United States of America
| | - Stephen Naylor
- ReNeuroGen L.L.C. Milwaukee, Elm Grove, Wisconsin, United States of America
| | - Martin J. Hessner
- Children’s Research Institute, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America,Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
| | - G. Ganesh Konduri
- Children’s Research Institute, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America,Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America
| | - Ru-Jeng Teng
- Children’s Research Institute, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America,Department of Pediatrics, Medical College of Wisconsin, Wauwatosa, Wisconsin, United States of America,* E-mail:
| |
Collapse
|
6
|
Yu S, Kong L, Gu L, Zhu Y, Liu X, Sang Y, Wang Q, Wang S, Zhang D, Cao H, Tao F, Liu K. Typical antibiotic exposure and dysglycemia risk in an elderly Chinese population. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:59701-59711. [PMID: 35394631 DOI: 10.1007/s11356-022-20056-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/29/2022] [Indexed: 06/14/2023]
Abstract
Studies examined the connection between antibiotic exposure in urine and dysglycemia risk (including prediabetes and diabetes) in the elderly were limited. Multiple linear regression, binary logistic regression, restricted cubic splines (RCS), and stratified analysis were applied to analyze the relationship between antibiotic exposure and dysglycemia risk. We observed that sulfaclozine exposure 0.07 (95% confidence interval [CI]: 0.01-0.23) significantly increased fasting blood glucose (FBG) level. By mechanism, usage, and antimicrobial action, sulfonamides 0.08 (95% CI: 0.06-0.36), veterinary antibiotics (VA) 0.07 (95% CI: 0.01-0.30), or bacteriostatic antibiotics 0.07 (95% CI: 0.02-0.29) significantly increased FBG level. Additionally, sulfaclozine exposure 1.54 (95% CI: 1.02-2.33) resulted in a higher dysglycemia risk, while doxycycline exposure 0.53 (95% CI: 0.30-0.95) resulted in a lower dysglycemia risk. By mechanism, usage, and antimicrobial action, sulfonamides 1.44 (95% CI: 1.02-2.04), VA 1.68 (95% CI: 1.21-2.35), or bacteriostatic antibiotics 1.40 (95% CI: 1.02-1.93) exposure had a higher dysglycemia risk. Taken together, exposure to sulfonamides, VA, especially sulfaclozine, was correlated with a higher dysglycemia risk in the elderly. Exposure to bacteriostatic antibiotics was associated with a higher dysglycemia risk in the female.
Collapse
Affiliation(s)
- Shuixin Yu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Li Kong
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Lvfen Gu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yitian Zhu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Xinji Liu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yanru Sang
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Qunan Wang
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Sufang Wang
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Dongmei Zhang
- School of Health Management, Anhui Medical University, Hefei, 230032, China
| | - Hongjuan Cao
- Lu'an Center of Disease Control and Prevention, Lu'an, Anhui, 237000, China
| | - Fangbiao Tao
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Kaiyong Liu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China.
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China.
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, No 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
7
|
Wu Y, Zong M, Wu H, He D, Li L, Zhang X, Zhao D, Li B. Dietary Advanced Glycation End-Products Affects the Progression of Early Diabetes by Intervening in Carbohydrate and Lipid Metabolism. Mol Nutr Food Res 2022; 66:e2200046. [PMID: 35355400 DOI: 10.1002/mnfr.202200046] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/01/2022] [Indexed: 12/18/2022]
Abstract
SCOPE Epidemiologic studies indicate significant contributions of thermally processed diets to the risk for diabetes and its related renal complications, but the mechanisms relating diet to disease remain unclear. This study evaluates the effects of the diet differ only in the content of advanced glycation end-products (AGEs) on early diabetes in Leprdb/db mice. METHODS AND RESULTS High AGEs diet (60 mg CML per kg protein) is fed to mice for 8 weeks. Dietary AGEs associated with diabetic features, including hyperglycemia, insulin resistance, and increased mRNA expression of renal chemokines, CCL3 and CXC3L1 are found. Untargeted metabolomics reveal that the high AGEs diet inhibits carbohydrate catabolism and promotes lipid anabolism. Additionally, the high AGEs diet alters the composition of the gut microbiota and indirectly affects the carbohydrate metabolism by altering the plasma levels of glyceraldehyde and pyruvate. However, switching to the lower AGEs diet can relieve most of the symptoms except microbiota composition. CONCLUSION The results indicate that dietary AGEs exposure intervenes in the development of diabetes through modulating the carbohydrate and lipid metabolism, and critically, switching to the lower AGEs diet arrested or reversed diabetes progression. A light-processing dietary intervention that helps to arrest early diabetes is suggested.
Collapse
Affiliation(s)
- Yi Wu
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, School of Food Science and Engineering, Engineering Research Center of Starch and Plant Protein Deep Processing, Ministry of Education, South China University of Technology, 381 Wushan Road, Tianhe District, Guangzhou, 510640, P. R. China
| | - Minhua Zong
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, School of Food Science and Engineering, Engineering Research Center of Starch and Plant Protein Deep Processing, Ministry of Education, South China University of Technology, 381 Wushan Road, Tianhe District, Guangzhou, 510640, P. R. China
| | - Hong Wu
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, School of Food Science and Engineering, Engineering Research Center of Starch and Plant Protein Deep Processing, Ministry of Education, South China University of Technology, 381 Wushan Road, Tianhe District, Guangzhou, 510640, P. R. China
| | - Dong He
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, No. 100 Waihuan Xi Road, Panyu District, Guangzhou, 510006, P. R. China
| | - Lin Li
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, School of Food Science and Engineering, Engineering Research Center of Starch and Plant Protein Deep Processing, Ministry of Education, South China University of Technology, 381 Wushan Road, Tianhe District, Guangzhou, 510640, P. R. China.,School of Chemical Engineering and Energy Technology, Dongguan University of Technology, College Road 1, Dongguan, 523808, P. R. China
| | - Xia Zhang
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, School of Food Science and Engineering, Engineering Research Center of Starch and Plant Protein Deep Processing, Ministry of Education, South China University of Technology, 381 Wushan Road, Tianhe District, Guangzhou, 510640, P. R. China
| | - Di Zhao
- Key Laboratory of Meat Processing, MOA; Key Laboratory of Meat Processing and Quality Control, MOE; Jiang Synergetic Innovation Center of Meat Production, Processing and Quality Control, Nanjing Agricultural University, Nanjing, 210095, P. R. China
| | - Bing Li
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, School of Food Science and Engineering, Engineering Research Center of Starch and Plant Protein Deep Processing, Ministry of Education, South China University of Technology, 381 Wushan Road, Tianhe District, Guangzhou, 510640, P. R. China
| |
Collapse
|
8
|
Spisni E, Turroni S, Alvisi P, Spigarelli R, Azzinnari D, Ayala D, Imbesi V, Valerii MC. Nutraceuticals in the Modulation of the Intestinal Microbiota: Current Status and Future Directions. Front Pharmacol 2022; 13:841782. [PMID: 35370685 PMCID: PMC8971809 DOI: 10.3389/fphar.2022.841782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/23/2022] [Indexed: 12/12/2022] Open
Abstract
Pharmaceutical interest in the human intestinal microbiota has increased considerably, because of the increasing number of studies linking the human intestinal microbial ecology to an increasing number of non-communicable diseases. Many efforts at modulating the gut microbiota have been made using probiotics, prebiotics and recently postbiotics. However, there are other, still little-explored opportunities from a pharmaceutical point of view, which appear promising to obtain modifications of the microbiota structure and functions. This review summarizes all in vitro, in vivo and clinical studies demonstrating the possibility to positively modulate the intestinal microbiota by using probiotics, prebiotics, postbiotics, essential oils, fungus and officinal plants. For the future, clinical studies investigating the ability to impact the intestinal microbiota especially by using fungus, officinal and aromatic plants or their extracts are required. This knowledge could lead to effective microbiome modulations that might support the pharmacological therapy of most non-communicable diseases in a near future.
Collapse
Affiliation(s)
- Enzo Spisni
- Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
- *Correspondence: Enzo Spisni,
| | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Patrizia Alvisi
- Pediatric Gastroenterology Unit, Maggiore Hospital, Bologna, Italy
| | - Renato Spigarelli
- Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Demetrio Azzinnari
- Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | | | - Veronica Imbesi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Maria Chiara Valerii
- Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
9
|
Cabrera SM, Coren AT, Pant T, Ciecko AE, Jia S, Roethle MF, Simpson PM, Atkinson SN, Salzman NH, Chen YG, Hessner MJ. Probiotic normalization of systemic inflammation in siblings of type 1 diabetes patients: an open-label pilot study. Sci Rep 2022; 12:3306. [PMID: 35228584 PMCID: PMC8885673 DOI: 10.1038/s41598-022-07203-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 02/10/2022] [Indexed: 12/14/2022] Open
Abstract
The incidence of type 1 diabetes (T1D) has increased, coinciding with lifestyle changes that have likely altered the gut microbiota. Dysbiosis, gut barrier dysfunction, and elevated systemic inflammation consistent with microbial antigen exposure, have been associated with T1D susceptibility and progression. A 6-week, single-arm, open-label pilot trial was conducted to investigate whether daily multi-strain probiotic supplementation could reduce this familial inflammation in 25 unaffected siblings of T1D patients. Probiotic supplementation was well-tolerated as reflected by high participant adherence and no adverse events. Community alpha and beta diversity were not altered between the pre- and post-supplement stool samplings. However, LEfSe analyses identified post-supplement enrichment of the family Lachnospiraceae, producers of the anti-inflammatory short chain fatty acid butyrate. Systemic inflammation was measured by plasma-induced transcription and quantified with a gene ontology-based composite inflammatory index (I.I.com). Post-supplement I.I.com was significantly reduced and pathway analysis predicted inhibition of numerous inflammatory mediators and activation of IL10RA. Subjects with the greatest post-supplement reduction in I.I.com exhibited significantly lower CD4+ CD45RO+ (memory):CD4+ CD45RA+ (naïve) T-cell ratios after supplementation. Post-supplement IL-12p40, IL-13, IL-15, IL-18, CCL2, and CCL24 plasma levels were significantly reduced, while post-supplement butyrate levels trended 1.4-fold higher. Probiotic supplementation may modify T1D susceptibility and progression and warrants further study.
Collapse
|
10
|
Zhang X, Wang H, Xie C, Hu Z, Zhang Y, Peng S, He Y, Kang J, Gao H, Yuan H, Liu Y, Fan G. Shenqi compound ameliorates type-2 diabetes mellitus by modulating the gut microbiota and metabolites. J Chromatogr B Analyt Technol Biomed Life Sci 2022; 1194:123189. [PMID: 35219959 DOI: 10.1016/j.jchromb.2022.123189] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/25/2022] [Accepted: 02/22/2022] [Indexed: 11/26/2022]
Abstract
The gut microbiota (GM) and metabolites are important factors in mediating the development of type-2 diabetes mellitus (T2DM). An imbalance in the gut microbiota and metabolites can disrupt the function of the intestinal barrier, cause changes in the permeability of the intestinal mucosa and promote the immune inflammatory response, thereby aggravating the fluctuation of blood glucose level and promoting the occurrence and development of the chronic complications of DM. Manipulating the GM and metabolites is a promising therapeutic intervention and is being studied extensively. Shenqi compound (SQC) is a traditional Chinese medicine formulation, which has been widely used to improve T2DM. Studies have demonstrated that SQC can reduce glycemic variability, alleviate the inflammatory response, etc. However, its underlying mechanism remains unknown. Therefore, in this experiment, We administered SQC to Goto-Kakizaki (GK) rats and evaluated its effect on blood glucose homeostasis and the intestinal mucosal barrier. We identified the profiles of the GM and metabolites with the aid of 16S rDNA gene sequencing and non-target metabolomics analysis. It showed that SQC intervention could reduce glycemic variability, regulate serum levels of glucagon and insulin, and improve injury to the intestinal mucosal barrier of GK rats. In the gut, the ratio of bacteria of the phyla Bacteroidetes/Firmicutes could be improved after SQC intervention. SQC also regulated the relative abundance of Prevotellaceae, Butyricimonas, Bacteroides, Blautia, Roseburia, Lactobacillus, and Rothia. We found out that expression of 40 metabolites was significantly improved after SQC intervention. Further analyses of metabolic pathways indicated that the therapeutic effect of SQC might be related predominantly to its ability to improve gluconeogenesis/glycolysis, amino acid metabolism, lipid metabolism, citrate cycle, and butanoate metabolism. These results suggest that SQC may exert a beneficial role in T2DM by modulating the GM and metabolites in different pathways.
Collapse
Affiliation(s)
- Xiyu Zhang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Heting Wang
- Department of Traditional Chinese Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Chunguang Xie
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhipeng Hu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuan Zhang
- First Affiliated Hospital of Army Medical University, Chongqing, China
| | - Sihan Peng
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuchi He
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jian Kang
- Department of Anorectal, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hong Gao
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Haipo Yuan
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ya Liu
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Gang Fan
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
11
|
Sargin P, Roethle MF, Jia S, Pant T, Ciecko AE, Atkinson SN, Salzman NH, Teng RJ, Chen YG, Cabrera SM, Hessner MJ. Lactiplantibacillus plantarum 299v supplementation modulates β-cell ER stress and antioxidative defense pathways and prevents type 1 diabetes in gluten-free BioBreeding rats. Gut Microbes 2022; 14:2136467. [PMID: 36261888 PMCID: PMC9586621 DOI: 10.1080/19490976.2022.2136467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 10/03/2022] [Indexed: 02/04/2023] Open
Abstract
The increasing incidence of Type 1 diabetes has coincided with the emergence of the low-fiber, high-gluten Western diet and other environmental factors linked to dysbiosis. Since Lactiplantibacillus plantarum 299 v (Lp299v) supplementation improves gut barrier function and reduces systemic inflammation, we studied its effects in spontaneously diabetic DRlyp/lyp rats provided a normal cereal diet (ND) or a gluten-free hydrolyzed casein diet (HCD). All rats provided ND developed diabetes (62.5±7.7 days); combining ND with Lp299v did not improve survival. Diabetes was delayed by HCD (72.2±9.4 days, p = .01) and further delayed by HCD+Lp299v (84.9±14.3 days, p < .001). HCD+Lp299v pups exhibited increased plasma propionate and butyrate levels, which correlated with enriched fecal Bifidobacteriaceae and Clostridiales taxa. Islet transcriptomic and histologic analyses at 40-days of age revealed that rats fed HCD expressed an autophagy profile, while those provided HCD+Lp299v expressed ER-associated protein degradation (ERAD) and antioxidative defense pathways, including Nrf2. Exposing insulinoma cells to propionate and butyrate promoted the antioxidative defense response but did not recapitulate the HCD+Lp299v islet ERAD transcriptomic profile. Here, both diet and microbiota influenced diabetes susceptibility. Moreover, Lp299v supplement modulated antioxidative defense and ER stress responses in β-cells, potentially offering a new therapeutic direction to thwart diabetes progression and preserve insulin secretion.
Collapse
Affiliation(s)
- Pinar Sargin
- The Max McGee Research Center for Juvenile Diabetes, Children’s Research Institute of Children’s Hospital of Wisconsin, Milwaukee, WI, USA
- Department of Pediatrics, Division of Endocrinology, the Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mark F. Roethle
- The Max McGee Research Center for Juvenile Diabetes, Children’s Research Institute of Children’s Hospital of Wisconsin, Milwaukee, WI, USA
- Department of Pediatrics, Division of Endocrinology, the Medical College of Wisconsin, Milwaukee, WI, USA
| | - Shuang Jia
- The Max McGee Research Center for Juvenile Diabetes, Children’s Research Institute of Children’s Hospital of Wisconsin, Milwaukee, WI, USA
- Department of Pediatrics, Division of Endocrinology, the Medical College of Wisconsin, Milwaukee, WI, USA
| | - Tarun Pant
- The Max McGee Research Center for Juvenile Diabetes, Children’s Research Institute of Children’s Hospital of Wisconsin, Milwaukee, WI, USA
- Department of Pediatrics, Division of Endocrinology, the Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ashley E. Ciecko
- The Max McGee Research Center for Juvenile Diabetes, Children’s Research Institute of Children’s Hospital of Wisconsin, Milwaukee, WI, USA
- Department of Pediatrics, Division of Endocrinology, the Medical College of Wisconsin, Milwaukee, WI, USA
| | - Samantha N. Atkinson
- Center for Microbiome Research, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Nita H. Salzman
- Center for Microbiome Research, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Pediatrics, Division of Gastroenterology, the Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ru-Jeng Teng
- Department of Pediatrics, Division of Neonatology, the Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yi-Guang Chen
- The Max McGee Research Center for Juvenile Diabetes, Children’s Research Institute of Children’s Hospital of Wisconsin, Milwaukee, WI, USA
- Department of Pediatrics, Division of Endocrinology, the Medical College of Wisconsin, Milwaukee, WI, USA
| | - Susanne M. Cabrera
- The Max McGee Research Center for Juvenile Diabetes, Children’s Research Institute of Children’s Hospital of Wisconsin, Milwaukee, WI, USA
- Department of Pediatrics, Division of Endocrinology, the Medical College of Wisconsin, Milwaukee, WI, USA
| | - Martin J. Hessner
- The Max McGee Research Center for Juvenile Diabetes, Children’s Research Institute of Children’s Hospital of Wisconsin, Milwaukee, WI, USA
- Department of Pediatrics, Division of Endocrinology, the Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
12
|
Wang S, Tan Q, Hou Y, Dou H. Emerging Roles of Myeloid-Derived Suppressor Cells in Diabetes. Front Pharmacol 2021; 12:798320. [PMID: 34975496 PMCID: PMC8716856 DOI: 10.3389/fphar.2021.798320] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/01/2021] [Indexed: 12/12/2022] Open
Abstract
Diabetes is a syndrome characterized by hyperglycemia with or without insulin resistance. Its etiology is attributed to the combined action of genes, environment and immune cells. Myeloid-derived suppressor cell (MDSC) is a heterogeneous population of immature cells with immunosuppressive ability. In recent years, different studies have debated the quantity, activity changes and roles of MDSC in the diabetic microenvironment. However, the emerging roles of MDSC have not been fully documented with regard to their interactions with diabetes. Here, the manifestations of MDSC and their subsets are reviewed with regard to the incidence of diabetes and diabetic complications. The possible drugs targeting MDSC are discussed with regard to their potential of treating diabetes. We believe that understanding MDSC will offer opportunities to explain pathological characteristics of different diabetes. MDSC also will be used for personalized immunotherapy of diabetes.
Collapse
Affiliation(s)
- Shiqi Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Qian Tan
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
| | - Huan Dou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
| |
Collapse
|
13
|
Tian J, Zhao Y, Wang L, Li L. Role of TLR4/MyD88/NF-κB signaling in heart and liver-related complications in a rat model of type 2 diabetes mellitus. J Int Med Res 2021; 49:300060521997590. [PMID: 33787393 PMCID: PMC8020098 DOI: 10.1177/0300060521997590] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Aims To analyze expression of members of the Toll-like receptor (TLR)4/myeloid
differentiation primary response 88 (MyD88)/nuclear factor (NF)-κB signaling
pathway in the heart and liver in a rat model of type 2 diabetes mellitus
(T2DM). Our overall goal was to understand the underlying pathophysiological
mechanisms. Methods We measured fasting blood glucose (FBG) and insulin (FINS) in a rat model of
T2DM. Expression of members of the TLR4/MyD88/NF-κB signaling pathway as
well as downstream cytokines was investigated. Levels of mRNA and protein
were assessed using quantitative real-time polymerase chain reaction and
western blotting, respectively. Protein content of tissue homogenates was
assessed using enzyme-linked immunosorbent assays. Results Diabetic rats had lower body weights, higher FBG, higher FINS, and higher
intraperitoneal glucose tolerance than normal rats. In addition, biochemical
indicators related to heart and liver function were elevated in diabetic
rats compared with normal rats. TLR4 and MyD88 were involved in the
occurrence of T2DM as well as T2DM-related heart and liver complications.
TLR4 caused T2DM-related heart and liver complications through activation of
NF-κB. Conclusions TLR4/MyD88/NF-κB signaling induces production of tumor necrosis factor-α,
interleukin-6, and monocyte chemoattractant protein-1, leading to the heart-
and liver-related complications of T2DM.
Collapse
Affiliation(s)
- Jiajia Tian
- Department of Endocrinology, Weifang Yidu Central Hospital, Weifang, P.R. China
| | - Yanyan Zhao
- Department of Endocrinology, Weifang Yidu Central Hospital, Weifang, P.R. China
| | - Lingling Wang
- Department of Endocrinology, Weifang Yidu Central Hospital, Weifang, P.R. China
| | - Lin Li
- The PLA Rocket Force Characteristic Medical Center, Beijing, P.R. China
| |
Collapse
|
14
|
Docimo G, Cangiano A, Romano RM, Pignatelli MF, Offi C, Paglionico VA, Galdiero M, Donnarumma G, Nigro V, Esposito D, Rotondi M, Candela G, Pasquali D. The Human Microbiota in Endocrinology: Implications for Pathophysiology, Treatment, and Prognosis in Thyroid Diseases. Front Endocrinol (Lausanne) 2020; 11:586529. [PMID: 33343507 PMCID: PMC7746874 DOI: 10.3389/fendo.2020.586529] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/15/2020] [Indexed: 12/12/2022] Open
Abstract
The human microbiota is an integral component in the maintenance of health and of the immune system. Microbiome-wide association studies have found numerous diseases associated to dysbiosis. Studies are needed to move beyond correlations and begin to address causation. Autoimmune thyroid diseases (ATD) are one of the most common organ-specific autoimmune disorders with an increasing prevalence, higher than 5% worldwide. Most frequent manifestations of ATD are Hashimoto's thyroiditis and Graves' disease. The exact etiology of ATD remains unknown. Until now it is not clear whether bacterial infections can trigger ATD or modulate the efficacy of treatment and prognosis. The aim of our review is to characterize the microbiota and in ATD and to evaluate the impact of dysbiosis on treatment and prognosis. Moreover, variation of gut microbiome has been associated with thyroid cancer and benign nodules. Here we will characterize the microbioma in benign thyroid nodules, and papillary thyroid cancer to evaluate their implications in the pathophysiology and progression.
Collapse
Affiliation(s)
- Giovanni Docimo
- Division of Thyroid Surgery, Department of Medical and Advanced Surgical Sciences, University of Campania “Luigi Vanvitelli”, School of Medicine, Naples, Italy
| | - Angelo Cangiano
- Division of Thyroid Surgery, Department of Medical and Advanced Surgical Sciences, University of Campania “Luigi Vanvitelli”, School of Medicine, Naples, Italy
| | - Roberto Maria Romano
- Division of Thyroid Surgery, Department of Medical and Advanced Surgical Sciences, University of Campania “Luigi Vanvitelli”, School of Medicine, Naples, Italy
| | - Marcello Filograna Pignatelli
- Division of Thyroid Surgery, Department of Medical and Advanced Surgical Sciences, University of Campania “Luigi Vanvitelli”, School of Medicine, Naples, Italy
| | - Chiara Offi
- Division of Thyroid Surgery, Department of Medical and Advanced Surgical Sciences, University of Campania “Luigi Vanvitelli”, School of Medicine, Naples, Italy
| | - Vanda Amoresano Paglionico
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Marilena Galdiero
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Giovanna Donnarumma
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Vincenzo Nigro
- Department of Precision Medicine, University of Campania ”Luigi Vanvitelli”, Naples, Italy
| | - Daniela Esposito
- Department of Endocrinology, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mario Rotondi
- Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, University of Pavia, Pavia, Italy
| | - Giancarlo Candela
- Division of Thyroid Surgery, Department of Medical and Advanced Surgical Sciences, University of Campania “Luigi Vanvitelli”, School of Medicine, Naples, Italy
| | - Daniela Pasquali
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| |
Collapse
|
15
|
Liu Y, Lou X. Type 2 diabetes mellitus-related environmental factors and the gut microbiota: emerging evidence and challenges. Clinics (Sao Paulo) 2020; 75:e1277. [PMID: 31939557 PMCID: PMC6945290 DOI: 10.6061/clinics/2020/e1277] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 10/04/2019] [Indexed: 01/15/2023] Open
Abstract
The gut microbiota is a group of over 38 trillion bacterial cells in the human microbiota that plays an important role in the regulation of human metabolism through its symbiotic relationship with the host. Changes in the gut microbial ecosystem are associated with increased susceptibility to metabolic disease in humans. However, the composition of the gut microbiota in those with type 2 diabetes mellitus and in the pathogenesis of metabolic diseases is not well understood. This article reviews the relationship between environmental factors and the gut microbiota in individuals with type 2 diabetes mellitus. Finally, we discuss the goal of treating type 2 diabetes mellitus by modifying the gut microbiota and the challenges that remain in this area.
Collapse
Affiliation(s)
- Yanfen Liu
- Jinhua Municipal Central Hospital, Department of Endocrinology Jinhua, 321000, China
| | - Xueyong Lou
- Jinhua Municipal Central Hospital, Department of Endocrinology Jinhua, 321000, China
- *Corresponding author. E-mail:
| |
Collapse
|
16
|
Christen U, Kimmel R. Chemokines as Drivers of the Autoimmune Destruction in Type 1 Diabetes: Opportunity for Therapeutic Intervention in Consideration of an Optimal Treatment Schedule. Front Endocrinol (Lausanne) 2020; 11:591083. [PMID: 33193102 PMCID: PMC7604482 DOI: 10.3389/fendo.2020.591083] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/30/2020] [Indexed: 12/16/2022] Open
Abstract
Type 1 diabetes (T1D) is mainly precipitated by the destruction of insulin-producing β-cells in the pancreatic islets of Langerhans by autoaggressive T cells. The etiology of the disease is still not clear, but besides genetic predisposition the exposure to environmental triggers seems to play a major role. Virus infection of islets has been demonstrated in biopsies of T1D patients, but there is still no firm proof that such an infection indeed results in islet-specific autoimmunity. However, virus infection results in a local inflammation with expression of inflammatory factors, such as cytokines and chemokines that attract and activate immune cells, including potential autoreactive T cells. Many chemokines have been found to be elevated in the serum and expressed by islet cells of T1D patients. In mouse models, it has been demonstrated that β-cells express chemokines involved in the initial recruitment of immune cells to the islets. The bulk load of chemokines is however released by the infiltrating immune cells that also express multiple chemokine receptors. The result is a mutual attraction of antigen-presenting cells and effector immune cells in the local islet microenvironment. Although there is a considerable redundancy within the chemokine ligand-receptor network, a few chemokines, such as CXCL10, seem to play a key role in the T1D pathogenesis. Studies with neutralizing antibodies and investigations in chemokine-deficient mice demonstrated that interfering with certain chemokine ligand-receptor axes might also ameliorate human T1D. However, one important aspect of such a treatment is the time of administration. Blockade of the recruitment of immune cells to the site of autoimmune destruction might not be effective when the disease process is already ongoing. By that time, autoaggressive cells have already arrived in the islet microenvironment and a blockade of migration might even hold them in place leading to accelerated destruction. Thus, an anti-chemokine therapy makes most sense in situations where the cells have not yet migrated to the islets. Such situations include treatment of patients at risk already carrying islet-antigen autoantibodies but are not yet diabetic, islet transplantation recipients, and patients that have undergone a T cell reset as occurring after anti-CD3 antibody treatment.
Collapse
|
17
|
Pellicano C, Leodori G, Innocenti GP, Gigante A, Rosato E. Microbiome, Autoimmune Diseases and HIV Infection: Friends or Foes? Nutrients 2019; 11:E2629. [PMID: 31684052 PMCID: PMC6893726 DOI: 10.3390/nu11112629] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/25/2019] [Accepted: 10/30/2019] [Indexed: 02/07/2023] Open
Abstract
Several studies highlighted the importance of the interaction between microbiota and the immune system in the development and maintenance of the homeostasis of the human organism. Dysbiosis is associated with proinflammatory and pathological state-like metabolic diseases, autoimmune diseases and HIV infection. In this review, we discuss the current understanding of the possible role of dysbiosis in triggering and/or exacerbating symptoms of autoimmune diseases and HIV infection. There are no data about the influence of the microbiome on the development of autoimmune diseases during HIV infection. We can hypothesize that untreated patients may be more susceptible to the development of autoimmune diseases, due to the presence of dysbiosis. Eubiosis, re-established by probiotic administration, can be used to reduce triggers for autoimmune diseases in untreated HIV patients, although clinical studies are needed to evaluate the role of the microbiome in autoimmune diseases in HIV patients.
Collapse
Affiliation(s)
- Chiara Pellicano
- Department of Translational and Precision Medicine-Scleroderma Unit, Sapienza University of Rome, 00185 Rome, Italy.
| | - Giorgia Leodori
- Department of Translational and Precision Medicine-Scleroderma Unit, Sapienza University of Rome, 00185 Rome, Italy.
| | | | - Antonietta Gigante
- Department of Translational and Precision Medicine-Scleroderma Unit, Sapienza University of Rome, 00185 Rome, Italy.
| | - Edoardo Rosato
- Department of Translational and Precision Medicine-Scleroderma Unit, Sapienza University of Rome, 00185 Rome, Italy.
| |
Collapse
|
18
|
Gianchecchi E, Fierabracci A. Recent Advances on Microbiota Involvement in the Pathogenesis of Autoimmunity. Int J Mol Sci 2019; 20:E283. [PMID: 30642013 PMCID: PMC6359510 DOI: 10.3390/ijms20020283] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/03/2019] [Accepted: 01/07/2019] [Indexed: 02/07/2023] Open
Abstract
Autoimmune disorders derive from genetic, stochastic, and environmental factors that all together interact in genetically predisposed individuals. The impact of an imbalanced gut microbiome in the pathogenesis of autoimmunity has been suggested by an increasing amount of experimental evidence, both in animal models and humans. Several physiological mechanisms, including the establishment of immune homeostasis, are influenced by commensal microbiota in the gut. An altered microbiota composition produces effects in the gut immune system, including defective tolerance to food antigens, intestinal inflammation, and enhanced gut permeability. In particular, early findings reported differences in the intestinal microbiome of subjects affected by several autoimmune conditions, including prediabetes or overt disease compared to healthy individuals. The present review focuses on microbiota-host homeostasis, its alterations, factors that influence its composition, and putative involvement in the development of autoimmune disorders. In the light of the existing literature, future studies are necessary to clarify the role played by microbiota modifications in the processes that cause enhanced gut permeability and molecular mechanisms responsible for autoimmunity onset.
Collapse
Affiliation(s)
- Elena Gianchecchi
- Infectivology and Clinical Trials Research Department, Children's Hospital Bambino Gesù, Viale San Paolo 15, 00146 Rome, Italy.
- VisMederi s.r.l., Strada del Petriccio e Belriguardo, 35, 53100 Siena, Italy.
| | - Alessandra Fierabracci
- Infectivology and Clinical Trials Research Department, Children's Hospital Bambino Gesù, Viale San Paolo 15, 00146 Rome, Italy.
| |
Collapse
|
19
|
Crookshank JA, Serrano D, Wang GS, Patrick C, Morgan BS, Paré MF, Scott FW. Changes in insulin, glucagon and ER stress precede immune activation in type 1 diabetes. J Endocrinol 2018; 239:181-195. [PMID: 30139929 DOI: 10.1530/joe-18-0328] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 08/06/2018] [Indexed: 12/11/2022]
Abstract
It is unknown whether there is a gene signature in pancreas which is associated with type 1 diabetes (T1D). We performed partial pancreatectomies on 30-day preinsulitic, diabetes-prone BioBreeding (BBdp) rats to prospectively identify factors involved in early prediabetes. Microarrays of the biopsies revealed downregulation of endoplasmic reticulum (ER) stress, metabolism and apoptosis. Based on these results, additional investigations compared gene expression in control (BBc) and BBdp rats age ~8, 30 and 60 days using RT-qPCR. Neonates had increased ER stress gene expression in pancreas. This was associated with decreased insulin, cleaved caspase-3 and Ins1 whereas Gcg and Pcsk2 were increased. The increase in ER stress was not sustained at 30 days and decreased by 60 days. In parallel, the liver gene profile showed a similar signature in neonates but with an early decrease of the unfolded protein response (UPR) at 30 days. This suggested that changes in the liver precede those in the pancreas. Tnf and Il1b expression was increased in BBdp pancreas in association with increased caspase-1, cleaved caspase-3 and decreased proinsulin area. Glucagon area was increased in both 30-day and 60-day BBdp rats. Increased colocalization of BIP and proinsulin was observed at 60 days in the pancreas, suggesting insulin-related ER dysfunction. We propose that dysregulated metabolism leads to ER stress in neonatal rats long before insulitis, creating a microenvironment in both pancreas and liver that promotes autoimmunity.
Collapse
Affiliation(s)
- Jennifer A Crookshank
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Daniel Serrano
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Gen-Sheng Wang
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Christopher Patrick
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Baylie S Morgan
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Biomedical Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Marie-France Paré
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Fraser W Scott
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
- Department of Medicine, The Ottawa Hospital, Ottawa, Ontario, Canada
| |
Collapse
|
20
|
Corbin KD, Driscoll KA, Pratley RE, Smith SR, Maahs DM, Mayer-Davis EJ. Obesity in Type 1 Diabetes: Pathophysiology, Clinical Impact, and Mechanisms. Endocr Rev 2018; 39:629-663. [PMID: 30060120 DOI: 10.1210/er.2017-00191] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 06/21/2018] [Indexed: 02/07/2023]
Abstract
There has been an alarming increase in the prevalence of obesity in people with type 1 diabetes in recent years. Although obesity has long been recognized as a major risk factor for the development of type 2 diabetes and a catalyst for complications, much less is known about the role of obesity in the initiation and pathogenesis of type 1 diabetes. Emerging evidence suggests that obesity contributes to insulin resistance, dyslipidemia, and cardiometabolic complications in type 1 diabetes. Unique therapeutic strategies may be required to address these comorbidities within the context of intensive insulin therapy, which promotes weight gain. There is an urgent need for clinical guidelines for the prevention and management of obesity in type 1 diabetes. The development of these recommendations will require a transdisciplinary research strategy addressing metabolism, molecular mechanisms, lifestyle, neuropsychology, and novel therapeutics. In this review, the prevalence, clinical impact, energy balance physiology, and potential mechanisms of obesity in type 1 diabetes are described, with a special focus on the substantial gaps in knowledge in this field. Our goal is to provide a framework for the evidence base needed to develop type 1 diabetes-specific weight management recommendations that account for the competing outcomes of glycemic control and weight management.
Collapse
Affiliation(s)
- Karen D Corbin
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, Florida
| | - Kimberly A Driscoll
- Department of Pediatrics, School of Medicine, University of Colorado Denver, Aurora, Colorado.,Barbara Davis Center for Diabetes, Aurora, Colorado
| | - Richard E Pratley
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, Florida
| | - Steven R Smith
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, Florida
| | - David M Maahs
- Division of Pediatric Endocrinology, Department of Pediatrics, Stanford University, Stanford, California
| | - Elizabeth J Mayer-Davis
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | |
Collapse
|
21
|
Chhabra P, Spano AJ, Bowers D, Ren T, Moore DJ, Timko MP, Wu M, Brayman KL. Evidence for the Role of the Cecal Microbiome in Maintenance of Immune Regulation and Homeostasis. Ann Surg 2018; 268:541-549. [PMID: 29994931 DOI: 10.1097/sla.0000000000002930] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVE (S) Our objective was to investigate alterations in the cecal microbial composition during the development of type 1 diabetes (T1D) with or without IgM therapy, and correlate these alterations with the corresponding immune profile. METHODS (1) Female nonobese diabetic (NOD) mice treated with IgM or saline (n = 20/group) were divided into 5-week-old nondiabetic; 9 to 12-week-old prehyperglycemic stage-1; ≥13-week-old prehyperglycemic stage-2; and diabetic groups. 16S rRNA libraries were prepared from bacterial DNA and deep-sequenced. (2) New-onset diabetic mice were treated with IgM (200 μg on Days 1, 3, and 5) and their blood glucose monitored for 2 months. RESULTS Significant dysbiosis was observed in the cecal microbiome with the progression of T1D development. The alteration in microbiome composition was characterized by an increase in the bacteroidetes:firmicutes ratio. In contrast, IgM conserved normal bacteroidetes:firmicutes ratio and this effect was long-lasting. Furthermore, oral gavage using cecal content from IgM-treated mice significantly diminished the incidence of diabetes compared with controls, indicating that IgM specifically affected mucosa-associated microbes, and that the affect was causal and not an epiphenomenon. Also, regulatory immune cell populations (myeloid-derived suppressor cells and regulatory T cells) were expanded and insulin autoantibody production diminished in the IgM-treated mice. In addition, IgM therapy reversed hyperglycemia in 70% of new-onset diabetic mice (n = 10) and the mice remained normoglycemic for the entire post-treatment observation period. CONCLUSIONS The cecal microbiome appears to be important in maintaining immune homeostasis and normal immune responses.
Collapse
Affiliation(s)
- Preeti Chhabra
- Department of Surgery, University of Virginia, Charlottesville, VA
| | - Anthony J Spano
- Department of Biology, University of Virginia, Charlottesville, VA
| | - Daniel Bowers
- Department of Surgery, University of Virginia, Charlottesville, VA
| | - Tiantian Ren
- Department of Biology, University of Virginia, Charlottesville, VA
| | - Daniel J Moore
- Department of Pediatrics, Vanderbilt University, Nashville, TN
| | - Michael P Timko
- Department of Biology, University of Virginia, Charlottesville, VA
| | - Martin Wu
- Department of Biology, University of Virginia, Charlottesville, VA
| | | |
Collapse
|