1
|
La Merrill MA, Smith MT, McHale CM, Heindel JJ, Atlas E, Cave MC, Collier D, Guyton KZ, Koliwad S, Nadal A, Rhodes CJ, Sargis RM, Zeise L, Blumberg B. Consensus on the key characteristics of metabolism disruptors. Nat Rev Endocrinol 2025; 21:245-261. [PMID: 39613954 PMCID: PMC11916920 DOI: 10.1038/s41574-024-01059-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/28/2024] [Indexed: 12/01/2024]
Abstract
Metabolism-disrupting agents (MDAs) are chemical, infectious or physical agents that increase the risk of metabolic disorders. Examples include pharmaceuticals, such as antidepressants, and environmental agents, such as bisphenol A. Various types of studies can provide evidence to identify MDAs, yet a systematic method is needed to integrate these data to help to identify such hazards. Inspired by work to improve hazard identification of carcinogens using key characteristics (KCs), we developed 12 KCs of MDAs based on our knowledge of processes underlying metabolic diseases and the effects of their causal agents: (1) alters function of the endocrine pancreas; (2) impairs function of adipose tissue; (3) alters nervous system control of metabolic function; (4) promotes insulin resistance; (5) disrupts metabolic signalling pathways; (6) alters development and fate of metabolic cell types; (7) alters energy homeostasis; (8) causes inappropriate nutrient handling and partitioning; (9) promotes chronic inflammation and immune dysregulation in metabolic tissues; (10) disrupts gastrointestinal tract function; (11) induces cellular stress pathways; and (12) disrupts circadian rhythms. In this Consensus Statement, we present the logic that revealed the KCs of MDAs and highlight evidence that supports the identification of KCs. We use chemical, infectious and physical agents as examples to illustrate how the KCs can be used to organize and use mechanistic data to help to identify MDAs.
Collapse
Affiliation(s)
- Michele A La Merrill
- Department of Environmental Toxicology, University of California, Davis, CA, USA.
| | - Martyn T Smith
- School of Public Health, University of California, Berkeley, CA, USA
| | - Cliona M McHale
- School of Public Health, University of California, Berkeley, CA, USA
| | - Jerrold J Heindel
- Healthy Environment and Endocrine Disruptor Strategies, Environmental Health Sciences, Bozeman, MT, USA
| | - Ella Atlas
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Matthew C Cave
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY, USA
| | - David Collier
- Department of Pediatrics, East Carolina University, Greenville, NC, USA
| | - Kathryn Z Guyton
- Board on Environmental Studies and Toxicology, National Academies of Sciences, Engineering, and Medicine, Washington, DC, USA
| | - Suneil Koliwad
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Angel Nadal
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), CIBERDEM, Miguel Hernandez University of Elche, Elche, Spain
| | - Christopher J Rhodes
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Robert M Sargis
- Division of Endocrinology, Diabetes and Metabolism, The University of Illinois at Chicago, Chicago, IL, USA
| | - Lauren Zeise
- Office of the Director, Office of Environmental Health Hazard Assessment of the California Environmental Protection Agency, Sacramento, CA, USA
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
2
|
Medina CK, Aykut B. Gut Microbial Dysbiosis and Implications in Solid Organ Transplantation. Biomedicines 2024; 12:2792. [PMID: 39767699 PMCID: PMC11673786 DOI: 10.3390/biomedicines12122792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/29/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
The gut microbiome has been shown to play a significant role in solid organ transplantation, potentially influencing graft function and patient outcomes. Dysbiosis, characterized by reduced microbial diversity and an increase in pathogenic taxa, has been linked to higher incidences of allograft rejection, graft dysfunction, and post-transplant mortality. Several studies suggest that the gut microbiome might be able to serve as both a biomarker and a therapeutic target, potentially guiding personalized immunosuppressive therapies and other interventions to improve outcomes after solid organ transplantation. As summarized in this review, clinical studies have shown that specific microbial shifts correlate with adverse outcomes, including acute rejection and chronic allograft dysfunction. As research surrounding the relationship between the gut microbiome and solid organ transplant progresses, the integration of microbial analysis into clinical practice has the potential to revolutionize post-transplant care, offering new avenues to improve graft survival and patient quality of life. This review aims to provide a comprehensive overview of the relationship between gut microbial dysbiosis and transplantation outcomes, emphasizing the impact on kidney, liver, lung, and heart transplant recipients.
Collapse
Affiliation(s)
| | - Berk Aykut
- Department of Surgery, Duke University, Durham, NC 27710, USA
| |
Collapse
|
3
|
Mazhar MU, Naz S, Khan JZ, Azam S, Ghazanfar S, Tipu MK. Protective potential of Bacillus subtilis (NMCC-path-14) against extraarticular manifestations during acute and sub-acute phase of arthritis using mice model. Biochem Biophys Res Commun 2024; 733:150708. [PMID: 39298918 DOI: 10.1016/j.bbrc.2024.150708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/29/2024] [Accepted: 09/14/2024] [Indexed: 09/22/2024]
Abstract
Extra-articular manifestations (EAM), which are associated with rheumatoid arthritis (RA), affect the quality of life of patients and are one of the critical causes of early mortality. This study was aimed at investigating whether Bacillus subtilis NMCC-path-14 (1 × 108 CFU/animal/day) could serve as a valuable therapeutic agent in managing EAM using complete Freund's adjuvant (CFA) induced arthritis during acute and sub-acute phases. Arthritis was induced using intra-dermal administration of CFA in the right hind paw of mice on day 1. Dexamethasone (Dexa) (5 mg/kg/day/animal) was used as a standard treatment. Animals in Dexa and Bacillus subtilis concurrent treatment (BS-CT) received treatments on day 1. The Bacillus subtilis pre-treatment (BS-PT) group received a probiotic dose 7 days before arthritis induction. Parameters like body weight, relative organ weight, colon length, hematology, serum biochemistry, antioxidant capacity, and histopathology of liver, kidney, spleen, colon, stress-related behavioral changes, and cortisol levels were evaluated on days 7 (acute) and 14 (sub-acute). Dexa failed to manage the EAM in arthritic mice and instead exacerbated them. On the other hand, B. subtilis NMCC-path-14 significantly declined EAM with no notable side effects, highlighting its safety and effectiveness. The current data show that B. subtilis NMCC-path-14 may be an alternative option for arthritis treatment that can reduce systemic symptoms associated with arthritis. More studies are required to comprehend the underlying mechanisms of mitigating the EAM by B. subtilis NMCC-path-14.
Collapse
Affiliation(s)
- Muhammad Usama Mazhar
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Sadaf Naz
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Jehan Zeb Khan
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Shahzad Azam
- Department of Pathology, Fazaia Medical College, Air University, Islamabad, Pakistan.
| | - Shakira Ghazanfar
- National Institute for Genomics and Advanced Biotechnology (NIGAB), National Agricultural Research Centre (NARC), Islamabad, Pakistan.
| | - Muhammad Khalid Tipu
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| |
Collapse
|
4
|
Green GBH, Cox-Holmes AN, Backan O, Valbak O, Potier ACE, Chen D, Morrow CD, Willey CD, McFarland BC. Exploring Gut Microbiota Alterations with Trimethoprim-Sulfamethoxazole and Dexamethasone in a Humanized Microbiome Mouse Model. Microorganisms 2024; 12:1015. [PMID: 38792844 PMCID: PMC11124107 DOI: 10.3390/microorganisms12051015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Along with the standard therapies for glioblastoma, patients are commonly prescribed trimethoprim-sulfamethoxazole (TMP-SMX) and dexamethasone for preventing infections and reducing cerebral edema, respectively. Because the gut microbiota impacts the efficacy of cancer therapies, it is important to understand how these medications impact the gut microbiota of patients. Using mice that have been colonized with human microbiota, this study sought to examine how TMP-SMX and dexamethasone affect the gut microbiome. Two lines of humanized microbiota (HuM) Rag1-/- mice, HuM1Rag and HuM2Rag, were treated with either TMP-SMX or dexamethasone via oral gavage once a day for a week. Fecal samples were collected pre-treatment (pre-txt), one week after treatment initiation (1 wk post txt), and three weeks post-treatment (3 wk post txt), and bacterial DNA was analyzed using 16S rRNA-sequencing. The HuM1Rag mice treated with TMP-SMX had significant shifts in alpha diversity, beta diversity, and functional pathways at all time points, whereas in the HuM2Rag mice, it resulted in minimal changes in the microbiome. Likewise, dexamethasone treatment resulted in significant changes in the microbiome of the HuM1Rag mice, whereas the microbiome of the HuM2Rag mice was mostly unaffected. The results of our study show that routine medications used during glioblastoma treatment can perturb gut microbiota, with some microbiome compositions being more sensitive than others, and these treatments could potentially affect the overall efficacy of standard-of-care therapy.
Collapse
Affiliation(s)
- George B. H. Green
- Department of Cell, Developmental and Integrative Biology, Birmingham, AL 35294, USA
| | - Alexis N. Cox-Holmes
- Department of Cell, Developmental and Integrative Biology, Birmingham, AL 35294, USA
| | - Olivia Backan
- Department of Cell, Developmental and Integrative Biology, Birmingham, AL 35294, USA
- Undergraduate Cancer Biology Program, Birmingham, AL 35294, USA
| | - Olivia Valbak
- Department of Cell, Developmental and Integrative Biology, Birmingham, AL 35294, USA
- Undergraduate Cancer Biology Program, Birmingham, AL 35294, USA
| | - Anna Claire E. Potier
- Department of Cell, Developmental and Integrative Biology, Birmingham, AL 35294, USA
- Undergraduate Cancer Biology Program, Birmingham, AL 35294, USA
| | | | - Casey D. Morrow
- Department of Cell, Developmental and Integrative Biology, Birmingham, AL 35294, USA
| | - Christopher D. Willey
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Braden C. McFarland
- Department of Cell, Developmental and Integrative Biology, Birmingham, AL 35294, USA
| |
Collapse
|
5
|
Dunn KA, MacDonald E, MacDonald T, Kulkarni K. Bacterial heat shock protein genes during induction chemotherapy in pediatric patients with acute lymphoblastic leukemia. Future Oncol 2024; 20:17-23. [PMID: 38189148 DOI: 10.2217/fon-2023-0263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024] Open
Abstract
Background: Heat shock proteins (HSP) protect cancer cells. Gastrointestinal bacteria contain HSP genes and can release extracellular vesicles which act as biological shuttles. Stress from treatment may result in a microbial community with more HSP genes, which could contribute to circulating HSP levels. Methods: The authors examined the abundance of five bacterial HSP genes pre-treatment and during induction in stool sequences from 30 pediatric acute lymphoblastic leukemia patients. Results: Decreased mean HTPG counts (p = 0.0024) pre-treatment versus induction were observed. During induction, HTPG, Shannon diversity and Bacteroidetes decreased (p = 7.5e-4; 1.1e-3; 8.6e-4), while DNAK and Firmicutes increased (p = 6.9e-3; 9.2e-4). Conclusion: Understanding microbial HSP gene community changes with treatment is the first step in determining if bacterial HSPs are important to the tumor microenvironment and leukemia treatment.
Collapse
Affiliation(s)
- Katherine A Dunn
- Department of Pediatrics, Division of Hematology Oncology, Izaak Walton Killam (IWK) Health, Halifax, NS, Canada
- Department of Biology, Dalhousie University, Halifax, NS, Canada
- Institute for Comparative Genomics, Dalhousie University, Halifax, NS, Canada
| | - Emma MacDonald
- Department of Pediatrics, Division of Hematology Oncology, Izaak Walton Killam (IWK) Health, Halifax, NS, Canada
| | - Tamara MacDonald
- Department of Pharmacy, IWK Health, Halifax, NS, Canada
- Faculty of Health Professions, Dalhousie University, Halifax, NS, Canada
| | - Ketan Kulkarni
- Department of Pediatrics, Division of Hematology Oncology, Izaak Walton Killam (IWK) Health, Halifax, NS, Canada
| |
Collapse
|
6
|
Qiu J, Cheng Y, Deng Y, Ren G, Wang J. Composition of gut microbiota involved in alleviation of dexamethasone-induced muscle atrophy by whey protein. NPJ Sci Food 2023; 7:58. [PMID: 37907516 PMCID: PMC10618183 DOI: 10.1038/s41538-023-00235-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 10/17/2023] [Indexed: 11/02/2023] Open
Abstract
Skeletal muscle atrophy is a condition associated with increased morbidity and mortality. While the concept of the gut-muscle axis has been proposed, the role of gut microbiota in dexamethasone (DEX)-induced skeletal muscle atrophy remains largely unknown, limiting its clinical applications. In this study, we found that administration of DEX caused a shift in the gut microbiota of mice, characterized by an increased ratio of Firmicutes/Bacteroidota and a reduction in alpha diversity. We also identified 480 new operational taxonomic units (OTUs), while 1168 specific OTUs were lost. Our Spearman correlation analysis revealed 28 key taxonomic genera of bacteria that were positively or negatively associated with skeletal muscle strength and weight (r: -0.881 to 0.845, p < 0.05). Moreover, supplementation with whey protein reshaped the gut microbiota structure in DEX-treated mice, making it more similar to that of the control group. Importantly, we further utilized a stepwise regression model to identify two enterotypes capable of predicting skeletal muscle function and weight. Notably, Ileibacterium and Lachnospiraceae_UCG-001 played significant roles in predicting both skeletal muscle function and weight. Our findings suggest that DEX causes shifts in the gut microbiota, which can be reversed by whey protein intervention. The enterotypes identified by our stepwise regression models predict muscle function and weight, underscoring the potential role of gut microbiota in modulating muscle atrophy and emphasizing the therapeutic opportunities of microbiota-altering interventions.
Collapse
Affiliation(s)
- JinLing Qiu
- Institute of Food and Nutrition Development, Ministry of Agriculture and Rural Affairs of the Reople's Republic of China, Beijing, China
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Yixing Cheng
- Institute of Food and Nutrition Development, Ministry of Agriculture and Rural Affairs of the Reople's Republic of China, Beijing, China
| | - Yang Deng
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Guangxu Ren
- Institute of Food and Nutrition Development, Ministry of Agriculture and Rural Affairs of the Reople's Republic of China, Beijing, China.
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong, China.
| | - Jiaqi Wang
- Institute of Food and Nutrition Development, Ministry of Agriculture and Rural Affairs of the Reople's Republic of China, Beijing, China.
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong, China.
| |
Collapse
|
7
|
Liu PY, Xia D, McGonigle K, Carroll AB, Chiango J, Scavello H, Martins R, Mehta S, Krespan E, Lunde E, LeVine D, Fellman CL, Goggs R, Beiting DP, Garden OA. Immune-mediated hematological disease in dogs is associated with alterations of the fecal microbiota: a pilot study. Anim Microbiome 2023; 5:46. [PMID: 37770990 PMCID: PMC10540429 DOI: 10.1186/s42523-023-00268-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 09/20/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND The dog is the most popular companion animal and is a valuable large animal model for several human diseases. Canine immune-mediated hematological diseases, including immune-mediated hemolytic anemia (IMHA) and immune thrombocytopenia (ITP), share many features in common with autoimmune hematological diseases of humans. The gut microbiome has been linked to systemic illness, but few studies have evaluated its association with immune-mediated hematological disease. To address this knowledge gap, 16S rRNA gene sequencing was used to profile the fecal microbiota of dogs with spontaneous IMHA and ITP at presentation and following successful treatment. In total, 21 affected and 13 healthy control dogs were included in the study. RESULTS IMHA/ITP is associated with remodeling of fecal microbiota, marked by decreased relative abundance of the spirochete Treponema spp., increased relative abundance of the pathobionts Clostridium septicum and Escherichia coli, and increased overall microbial diversity. Logistic regression analysis demonstrated that Treponema spp. were associated with decreased risk of IMHA/ITP (odds ratio [OR] 0.24-0.34), while Ruminococcaceae UCG-009 and Christensenellaceae R-7 group were associated with increased risk of disease (OR = 6.84 [95% CI 2-32.74] and 8.36 [95% CI 1.85-71.88] respectively). CONCLUSIONS This study demonstrates an association of immune-mediated hematological diseases in dogs with fecal dysbiosis, and points to specific bacterial genera as biomarkers of disease. Microbes identified as positive or negative risk factors for IMHA/ITP represent an area for future research as potential targets for new diagnostic assays and/or therapeutic applications.
Collapse
Affiliation(s)
- P-Y Liu
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, 804201, Taiwan
| | - D Xia
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
| | - K McGonigle
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, 3900 Spruce Street, Philadelphia, PA, 19104, USA
| | - A B Carroll
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, 3900 Spruce Street, Philadelphia, PA, 19104, USA
| | - J Chiango
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, 3900 Spruce Street, Philadelphia, PA, 19104, USA
| | - H Scavello
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, 3900 Spruce Street, Philadelphia, PA, 19104, USA
| | - R Martins
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, 3900 Spruce Street, Philadelphia, PA, 19104, USA
| | - S Mehta
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 380 South University Avenue, Philadelphia, 19104, USA
| | - E Krespan
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 380 South University Avenue, Philadelphia, 19104, USA
| | - E Lunde
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, 1809 South Riverside Drive, Ames, IA, 50011, USA
| | - D LeVine
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, 1809 South Riverside Drive, Ames, IA, 50011, USA
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, 1220 Wire Road, Auburn, AL, 36849, USA
| | - C L Fellman
- Department of Clinical Sciences, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, 01536, USA
| | - R Goggs
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, 930 Campus Road, Box 31, Ithaca, NY, 14853, USA
| | - D P Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 380 South University Avenue, Philadelphia, 19104, USA
| | - O A Garden
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, 3900 Spruce Street, Philadelphia, PA, 19104, USA.
- Dean's Office, School of Veterinary Medicine, Louisiana State University, Skip Bertman Drive, Baton Rouge, LA, 70803, USA.
| |
Collapse
|
8
|
Zhao J, Zhao F, Li X, Yuan J, Zhang K, Liu H, Wang Y. Multi-omics reveals the mechanisms underlying Lactiplantibacillus plantarum P8-mediated attenuation of oxidative stress in broilers challenged with dexamethasone. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2023; 14:281-302. [PMID: 37600839 PMCID: PMC10432922 DOI: 10.1016/j.aninu.2023.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 05/31/2023] [Accepted: 06/14/2023] [Indexed: 08/22/2023]
Abstract
Oxidative stress is a common phenomenon in poultry production. Several molecules, including antioxidant genes, miRNAs, and gut microbiota metabolites, have been reported to participate in redox regulation. Lactiplantibacillus plantarum P8 (P8) was shown to improve the antioxidant capacity of chickens, but the specific molecular mechanisms remain unclear. In this study, 400 broilers were allocated to 4 treatment groups: control diet (Con group), control diet + dexamethasone injection (DEX group), control diet containing 1 × 108 CFU/g P8 (P8 group), and control diet containing 1 × 108 CFU/g P8 + DEX injection (DEX_P8 group). Integrated analysis of the microbiome, metabolomics, and miRNAomics was conducted to investigate the roles of P8 in oxidative stress in broilers. Results demonstrated that P8 supplementation significantly improved growth performance, jejunal morphology, and antioxidant function in DEX-treated broilers. Analysis of the gut microbiota revealed a higher abundance of Barnesiella (P = 0.01) and Erysipelatoclostridium (P = 0.05) in the DEX_P8 group than in the DEX group. Functional prediction indicated that certain pathways, including the phenylacetate degradation pathway, were enriched in the DEX_P8 group compared to the DEX group. Metabolites in the cecal contents were distinct between the groups. P8 supplementation increased the content of metabolites with antioxidant capacity, e.g., urobilinogen (P < 0.01), and decreased that of metabolites related to oxidative stress, e.g., genistein (P < 0.01). Functional prediction indicated that metabolites that differed between the DEX_P8 and DEX groups were enriched in pathways including "tryptophan metabolism" and "primary bile acid biosynthesis". The miRNAomics analysis further showed that, compared to the DEX group, several miRNAs in the jejunum, such as gga-miR-21-3p (P = 0.03), were increased, whereas gga-miR-455-3p (P = 0.02) was decreased in the DEX_P8 group. The PI3K-Akt, Ras, and Rap1 signaling pathways were enriched in the DEX_P8 group compared to the DEX group through KEGG analysis. Correlation analysis revealed potential interactions between growth performance, oxidation/antioxidation, jejunal morphology, gut microbiota, cecal content metabolites, and jejunal miRNAs. Overall, our results indicate that P8 supplementation may improve the growth performance, jejunal morphology and antioxidant capacity of DEX-treated broilers by regulating gut microbiota, its metabolites, and intestinal miRNAs.
Collapse
Affiliation(s)
| | | | - Xuemin Li
- College of Animal Science and Technology, Qingdao Agricultural University, 266109, Qingdao, China
| | - Junmeng Yuan
- College of Animal Science and Technology, Qingdao Agricultural University, 266109, Qingdao, China
| | - Kai Zhang
- College of Animal Science and Technology, Qingdao Agricultural University, 266109, Qingdao, China
| | - Huawei Liu
- College of Animal Science and Technology, Qingdao Agricultural University, 266109, Qingdao, China
| | - Yang Wang
- College of Animal Science and Technology, Qingdao Agricultural University, 266109, Qingdao, China
| |
Collapse
|
9
|
Dallas JW, Warne RW. Captivity and Animal Microbiomes: Potential Roles of Microbiota for Influencing Animal Conservation. MICROBIAL ECOLOGY 2023; 85:820-838. [PMID: 35316343 DOI: 10.1007/s00248-022-01991-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 03/07/2022] [Indexed: 05/04/2023]
Abstract
During the ongoing biodiversity crisis, captive conservation and breeding programs offer a refuge for species to persist and provide source populations for reintroduction efforts. Unfortunately, captive animals are at a higher disease risk and reintroduction efforts remain largely unsuccessful. One potential factor in these outcomes is the host microbiota which includes a large diversity and abundance of bacteria, fungi, and viruses that play an essential role in host physiology. Relative to wild populations, the generalized pattern of gut and skin microbiomes in captivity are reduced alpha diversity and they exhibit a significant shift in community composition and/or structure which often correlates with various physiological maladies. Many conditions of captivity (antibiotic exposure, altered diet composition, homogenous environment, increased stress, and altered intraspecific interactions) likely lead to changes in the host-associated microbiome. To minimize the problems arising from captivity, efforts can be taken to manipulate microbial diversity and composition to be comparable with wild populations through methods such as increasing dietary diversity, exposure to natural environmental reservoirs, or probiotics. For individuals destined for reintroduction, these strategies can prime the microbiota to buffer against novel pathogens and changes in diet and improve reintroduction success. The microbiome is a critical component of animal physiology and its role in species conservation should be expanded and included in the repertoire of future management practices.
Collapse
Affiliation(s)
- Jason W Dallas
- Department of Biological Sciences, Southern Illinois University, 1125 Lincoln Drive, Carbondale, IL, 62901, USA.
| | - Robin W Warne
- Department of Biological Sciences, Southern Illinois University, 1125 Lincoln Drive, Carbondale, IL, 62901, USA
| |
Collapse
|
10
|
Couch CE, Neal WT, Herron CL, Kent ML, Schreck CB, Peterson JT. Gut microbiome composition associates with corticosteroid treatment, morbidity, and senescence in Chinook salmon (Oncorhynchus tshawytscha). Sci Rep 2023; 13:2567. [PMID: 36782001 PMCID: PMC9925776 DOI: 10.1038/s41598-023-29663-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Pacific salmon experience prolonged elevation in corticosteroid hormones during important life history events including migration, reproduction, and senescence. These periods of elevated corticosteroids correspond with changes to immunity and energy metabolism; therefore, fish may be particularly vulnerable to mortality at these times. Recent studies found that stress-induced cortisol release associated with microbial community shifts in salmonids, raising the question of how longer-term corticosteroid dynamics that accompany life history transitions affect salmonid microbiomes. In this work, we experimentally evaluated the relationships between gut microbiome composition, chronically elevated corticosteroids, and mortality in juvenile Chinook salmon (Oncorhynchus tshawytscha). We found that treatment with slow-release implants of the corticosteroids cortisol or dexamethasone resulted in changes to the gut microbiome. Morbidity was also associated with microbiome composition, suggesting that the gut microbiome reflects individual differences in susceptibility to opportunistic pathogens. Additionally, we analyzed a small number of samples from adult fish at various stages of senescence. Results from these samples suggest that microbiome composition associated with gut integrity, and that the microbial communities of corticosteroid treated juveniles shift in composition toward those of senescent adults. Overall, findings from this work indicate that the gut microbiome correlates with mortality risk during periods of chronic corticosteroid elevation.
Collapse
Affiliation(s)
- Claire E Couch
- Department of Fisheries, Wildlife, and Conservation Sciences, Oregon State University, Corvallis, OR, USA.
| | - William T Neal
- Department of Fisheries, Wildlife, and Conservation Sciences, Oregon State University, Corvallis, OR, USA
| | - Crystal L Herron
- Department of Fisheries, Wildlife, and Conservation Sciences, Oregon State University, Corvallis, OR, USA
| | - Michael L Kent
- Department of Microbiology, Oregon State University, Corvallis, OR, USA
- Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | - Carl B Schreck
- Department of Fisheries, Wildlife, and Conservation Sciences, Oregon State University, Corvallis, OR, USA
| | - James T Peterson
- Department of Fisheries, Wildlife, and Conservation Sciences, Oregon State University, Corvallis, OR, USA
- U.S. Geological Survey Oregon Cooperative Fish and Wildlife Research Unit, Corvallis, OR, USA
| |
Collapse
|
11
|
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: 2.3] [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
|
12
|
Efrain Molotla-Torres D, Mario Hernández-Soto L, Guzmán-Mejía F, Godínez-Victoria M, Elisa Drago-Serrano M, Félix Aguirre-Garrido J. Oral bovine lactoferrin modulation on fecal microbiota of mice underwent immobilization stress. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
13
|
Kozik V, Schwab M, Thiel S, Hellwig K, Rakers F, Dreiling M. Protocol for a Cross-Sectional Study: Effects of a Multiple Sclerosis Relapse Therapy With Methylprednisolone on Offspring Neurocognitive Development and Behavior (MS-Children). Front Neurol 2022; 13:830057. [PMID: 35557615 PMCID: PMC9087857 DOI: 10.3389/fneur.2022.830057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/17/2022] [Indexed: 11/21/2022] Open
Abstract
Introduction Multiple Sclerosis (MS) is the most common neuroimmunological disease in women of childbearing age. Current MS therapy consists of immunomodulatory relapse prevention with disease-modifying therapies (DMTs) and acute relapse therapy with the synthetic glucocorticoid (GC) methylprednisolone (MP). As most DMTs are not approved for use during pregnancy, treatment is usually discontinued, increasing the risk for relapses. While MP therapy during pregnancy is considered relatively save for the fetus, it may be detrimental for later cognitive and neuropsychiatric function. The underlying mechanism is thought to be an epigenetically mediated desensitization of GC receptors, the subsequent increase in stress sensitivity, and a GC-mediated impairment of brain development. The aim of this study is to investigate the associations of fetal MP exposure in the context of MS relapse therapy with later cognitive function, brain development, stress sensitivity, and behavior. Methods and Analysis Eighty children aged 8–18 years of mothers with MS will be recruited. Forty children, exposed to GC in utero will be compared to 40 children without fetal GC exposure. The intelligence quotient will serve as primary outcome. Secondary outcomes will include attention, motor development, emotional excitability, Attention-Deficit Hyperactivity Disorder-related symptoms, and behavioral difficulties. The Trier Social Stress Test will test stress sensitivity, EEG and MRI will assess functional and structural brain development. To determine underlying mechanisms, DNA methylation of the GC receptor gene and the H19/IGF2 locus and changes in the microbiome and the metabolome will be investigated. Primary and secondary outcomes will be analyzed using linear regression models. Time-variant outcomes of the stress test will be analyzed in two mixed linear models exploring overall activity and change from baseline. Ethics and Dissemination This study was approved by the participating institutions' ethics committees and results will be presented in accordance with the STROBE 2007 Statement. Trial Registration https://clinicaltrials.gov/ct2/show/NCT04832269?id=ZKSJ0130
Collapse
Affiliation(s)
- Valeska Kozik
- Department of Neurology, Jena University Hospital, Jena, Germany
- *Correspondence: Valeska Kozik
| | - Matthias Schwab
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - Sandra Thiel
- Department of Neurology, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Kerstin Hellwig
- Department of Neurology, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Florian Rakers
- Department of Neurology, Jena University Hospital, Jena, Germany
| | | |
Collapse
|
14
|
Huang R, Wu F, Zhou Q, Wei W, Yue J, Xiao B, Luo Z. Lactobacillus and intestinal diseases: mechanisms of action and clinical applications. Microbiol Res 2022; 260:127019. [DOI: 10.1016/j.micres.2022.127019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 12/12/2022]
|
15
|
Yang M, Zheng X, Wu Y, Zhang R, Yang Q, Yu Z, Liu J, Zha B, Gong Q, Yang B, Sun B, Zeng M. Preliminary Observation of the Changes in the Intestinal Flora of Patients With Graves’ Disease Before and After Methimazole Treatment. Front Cell Infect Microbiol 2022; 12:794711. [PMID: 35402292 PMCID: PMC8989835 DOI: 10.3389/fcimb.2022.794711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/14/2022] [Indexed: 11/23/2022] Open
Abstract
Immune dysfunction caused by environmental factors plays an important role in the development of Graves’ disease (GD), and environmental factors are closely related to the intestinal flora. Our previous study showed significant changes in the intestinal flora in GD patients compared with healthy volunteers. This study analyzed the relationships between changes in the intestinal flora, thyroid function and relevant thyroid antibodies in GD patients before and after methimazole treatment. The subjects were divided into the UGD group (18 newly diagnosed GD patients), the TGD group (10 GD patients with normal or approximately normal thyroid function after methimazole treatment) and the NC group (11 healthy volunteers). Their fresh stool samples were sent for 16S rRNA gene amplification and Illumina platform sequencing. The correlations of the relative abundance of Bifidobacterium with the levels of TRAb, TgAb and TPOAb in the NC group and the UGD group were analyzed. A total of 1,562,445 high-quality sequences were obtained. In the UGD group, the abundances of Bifidobacterium and Collinsella were higher than that in the NC group; Bacteroides abundance in the TGD group was higher than that in the NC group, while Prevotella and Dialister abundances were lower than that in the NC group; Prevotella and Collinsella abundances in the UGD group were higher than that in the TGD group. The predominant abundance distribution of Bifidobacteriaceae in the UGD group at the family level was superior to that in the NC group. The abundance of Bifidobacterium was positively correlated with the levels of TRAb, TgAb, and TPOAb. The biological diversity of the intestinal flora was reduced in GD patients. After methimazole treatment, the composition of the intestinal flora was significantly altered. The change in Bifidobacterium abundance was positively correlated with TRAb, TgAb and TPOAb, suggesting that it might be related to the immune mechanism of GD. The results of this study may deepen our understanding of the pathogenesis of GD and provide a new idea for the treatment of GD.
Collapse
Affiliation(s)
- Mengxue Yang
- Department of Endocrinology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
- *Correspondence: Mengxue Yang,
| | - Xiaodi Zheng
- Department of Endocrinology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Yueyue Wu
- Department of Endocrinology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Rui Zhang
- Department of Endocrinology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Qian Yang
- Department of Endocrinology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Zhiyan Yu
- Department of Endocrinology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Jun Liu
- Department of Endocrinology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Bingbing Zha
- Department of Endocrinology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Qihai Gong
- Key Laboratory of Basic Pharmacology of the Ministry of Education, Zunyi Medical University, Zunyi, China
- Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Bo Yang
- Department of Endocrinology, Zunyi Medical University, Zunyi, China
| | - Bowen Sun
- Department of Endocrinology, Zunyi Medical University, Zunyi, China
| | - Miao Zeng
- Department of Infectious Diseases I, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Hou J, Tang Y, Chen Y, Chen D. The Role of the Microbiota in Graves' Disease and Graves' Orbitopathy. Front Cell Infect Microbiol 2022; 11:739707. [PMID: 35004341 PMCID: PMC8727912 DOI: 10.3389/fcimb.2021.739707] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 12/01/2021] [Indexed: 02/05/2023] Open
Abstract
Graves' disease (GD) is a clinical syndrome with an enlarged and overactive thyroid gland, an accelerated heart rate, Graves' orbitopathy (GO), and pretibial myxedema (PTM). GO is the most common extrathyroidal complication of GD. GD/GO has a significant negative impact on the quality of life. GD is the most common systemic autoimmune disorder, mediated by autoantibodies to the thyroid-stimulating hormone receptor (TSHR). It is generally accepted that GD/GO results from complex interactions between genetic and environmental factors that lead to the loss of immune tolerance to thyroid antigens. However, the exact mechanism is still elusive. Systematic investigations into GD/GO animal models and clinical patients have provided important new insight into these disorders during the past 4 years. These studies suggested that gut microbiota may play an essential role in the pathogenesis of GD/GO. Antibiotic vancomycin can reduce disease severity, but fecal material transfer (FMT) from GD/GO patients exaggerates the disease in GD/GO mouse models. There are significant differences in microbiota composition between GD/GO patients and healthy controls. Lactobacillus, Prevotella, and Veillonella often increase in GD patients. The commonly used therapeutic agents for GD/GO can also affect the gut microbiota. Antigenic mimicry and the imbalance of T helper 17 cells (Th17)/regulatory T cells (Tregs) are the primary mechanisms proposed for dysbiosis in GD/GO. Interventions including antibiotics, probiotics, and diet modification that modulate the gut microbiota have been actively investigated in preclinical models and, to some extent, in clinical settings, such as probiotics (Bifidobacterium longum) and selenium supplements. Future studies will reveal molecular pathways linking gut and thyroid functions and how they impact orbital autoimmunity. Microbiota-targeting therapeutics will likely be an essential strategy in managing GD/GO in the coming years.
Collapse
Affiliation(s)
- Jueyu Hou
- The Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,The Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Yunjing Tang
- The Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,The Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Yongjiang Chen
- The School of Optometry and Vision Science, University of Waterloo, Waterloo, ON, Canada
| | - Danian Chen
- The Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,The Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
17
|
Xu X, Zhang Z. Sex- and age-specific variation of gut microbiota in Brandt's voles. PeerJ 2021; 9:e11434. [PMID: 34164232 PMCID: PMC8194415 DOI: 10.7717/peerj.11434] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 04/20/2021] [Indexed: 12/14/2022] Open
Abstract
Background Gut microbiota plays a key role in the survival and reproduction of wild animals which rely on microbiota to break down plant compounds for nutrients. As compared to laboratory animals, wild animals face much more threat of environmental changes (e.g. food shortages and risk of infection). Therefore, studying the gut microbiota of wild animals can help us better understand the mechanisms animals use to adapt to their environment. Methods We collected the feces of Brandt’s voles in the grassland, of three age groups (juvenile, adult and old), in both sexes. We studied the gut microbiota by 16S rRNA sequencing. Results The main members of gut microbiota in Brandt’s voles were Firmicutes, Bacteroidetes and Proteobacteria. As voles get older, the proportion of Firmicutes increased gradually, and the proportion of Bacteroides decreased gradually. The diversity of the microbiota of juveniles is lower, seems like there is still a lot of space for colonization, and there are large variations in the composition of the microbiome between individuals. In adulthood, the gut microbiota tends to be stable, and the diversity is highest. In adult, the abundances of Christensenellaceae and Peptococcus of female were significantly higher than male voles. Conclusions The gut microbiota of Brandt’s vole was influenced by sex and age, probably due to growth needs and hormone levels. Gut microbiota of wild animals were much influenced by their life-history reflected by their age and sex. Future studies will be directed to identify functions of these “wild microbiota” in regulating physiological or behavioral processes of wild animals in different life stage or sexes.
Collapse
Affiliation(s)
- Xiaoming Xu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, Beijing, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, Beijing, China
| | - Zhibin Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, Beijing, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, Beijing, China
| |
Collapse
|
18
|
Ericsson AC, Franklin CL. The gut microbiome of laboratory mice: considerations and best practices for translational research. Mamm Genome 2021; 32:239-250. [PMID: 33689000 PMCID: PMC8295156 DOI: 10.1007/s00335-021-09863-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 02/18/2021] [Indexed: 12/14/2022]
Abstract
Just as the gut microbiota (GM) is now recognized as an integral mediator of environmental influences on human physiology, susceptibility to disease, and response to pharmacological intervention, so too does the GM of laboratory mice affect the phenotype of research using mouse models. Multiple experimental factors have been shown to affect the composition of the GM in research mice, as well as the model phenotype, suggesting that the GM represents a major component in experimental reproducibility. Moreover, several recent studies suggest that manipulation of the GM of laboratory mice can substantially improve the predictive power or translatability of data generated in mouse models to the human conditions under investigation. This review provides readers with information related to these various factors and practices, and recommendations regarding methods by which issues with poor reproducibility or translatability can be transformed into discoveries.
Collapse
Affiliation(s)
- Aaron C Ericsson
- University of Missouri Metagenomics Center (MUMC), MU Mutant Mouse Resource and Research Center (MU MMRRC), Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA.
| | - Craig L Franklin
- University of Missouri Metagenomics Center (MUMC), MU Mutant Mouse Resource and Research Center (MU MMRRC), Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
19
|
Patrizz A, Dono A, Zorofchian S, Hines G, Takayasu T, Husein N, Otani Y, Arevalo O, Choi HA, Savarraj J, Tandon N, Ganesh BP, Kaur B, McCullough LD, Ballester LY, Esquenazi Y. Glioma and temozolomide induced alterations in gut microbiome. Sci Rep 2020; 10:21002. [PMID: 33273497 PMCID: PMC7713059 DOI: 10.1038/s41598-020-77919-w] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 11/11/2020] [Indexed: 12/19/2022] Open
Abstract
The gut microbiome is fundamental in neurogenesis processes. Alterations in microbial constituents promote inflammation and immunosuppression. Recently, in immune-oncology, specific microbial taxa have been described to enhance the effects of therapeutic modalities. However, the effects of microbial dysbiosis on glioma are still unknown. The aim of this study was to explore the effects of glioma development and Temozolomide (TMZ) on fecal microbiome in mice and humans. C57BL/6 mice were implanted with GL261/Sham and given TMZ/Saline. Fecal samples were collected longitudinally and analyzed by 16S rRNA sequencing. Fecal samples were collected from healthy controls as well as glioma patients at diagnosis, before and after chemoradiation. Compared to healthy controls, mice and glioma patients demonstrated significant differences in beta diversity, Firmicutes/Bacteroides (F/B) ratio, and increase of Verrucomicrobia phylum and Akkermansia genus. These changes were not observed following TMZ in mice. TMZ treatment in the non-tumor bearing mouse-model diminished the F/B ratio, increase Muribaculaceae family and decrease Ruminococcaceae family. Nevertheless, there were no changes in Verrucomicrobia/Akkermansia. Glioma development leads to gut dysbiosis in a mouse-model, which was not observed in the setting of TMZ. These findings seem translational to humans and warrant further study.
Collapse
Affiliation(s)
- Anthony Patrizz
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Antonio Dono
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA.,Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Soheil Zorofchian
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA.,Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Gabriella Hines
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Takeshi Takayasu
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA.,Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Nuruddin Husein
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Yoshihiro Otani
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Octavio Arevalo
- Department of Diagnostic and Interventional Imaging, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - H Alex Choi
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Jude Savarraj
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Nitin Tandon
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Bhanu P Ganesh
- Department of Neurology, The University of Texas Health Science Center At Houston, McGovern Medical School, Houston, TX, USA
| | - Balveen Kaur
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Louise D McCullough
- Department of Neurology, The University of Texas Health Science Center At Houston, McGovern Medical School, Houston, TX, USA
| | - Leomar Y Ballester
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA. .,Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA. .,Memorial Hermann Hospital-TMC, Houston, TX, USA. .,Department of Pathology & Laboratory Medicine and Department of Neurosurgery, The University of Texas Health Science Center at Houston - McGovern Medical School, 6431 Fannin Street, MSB 2.136, Houston, TX, 77030, USA.
| | - Yoshua Esquenazi
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA. .,Center for Precision Health, The University of Texas Health Science Center At Houston, McGovern Medical School, Houston, TX, USA. .,Memorial Hermann Hospital-TMC, Houston, TX, USA. .,Vivian L. Smith Department of Neurosurgery and Center for Precision Health, The University of Texas Health Science Center at Houston - McGovern Medical School, 6400 Fannin Street, Suite # 2800, Houston, TX, 77030, USA.
| |
Collapse
|
20
|
An G, Tang Y, Mo B, Ran M, He X, Bao J, Zhou Z. Characterization of a Murine Model for Encephalitozoon hellem Infection after Dexamethasone Immunosuppression. Microorganisms 2020; 8:E1891. [PMID: 33260440 PMCID: PMC7761425 DOI: 10.3390/microorganisms8121891] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 11/22/2020] [Accepted: 11/27/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Encephalitozoon hellem (E. hellem) belongs to a group of opportunistic pathogens called microsporidia. Microsporidia infection symptoms vary and include diarrhea, ocular disorders and systemic inflammations. Traditionally, immunodeficient animals were used to study microsporidia infection. To overcome the difficulties in maintenance and operation using immunodeficient mice, and to better mimic natural occurring microsporidia infection, this study aims to develop a pharmacologically immunosuppressed murine model of E. hellem infection. METHODS Wild-type C57BL/6 mice were immunosuppressed with dexamethasone (Dex) and then E. hellem spores were inoculated into the mice intraperitoneally. Control groups were the Dex-immunosuppressed but noninoculated mice, and the Dex-immunosuppressed then lipopolysaccharide (LPS)-treated mice. Mice body weights were monitored and all animals were sacrificed at the 15th day after inoculation. Tissue fragments and immune cells were collected and processed. RESULTS Histopathological analysis demonstrated that E. hellem inoculation resulted in a disseminated nonlethal infection. Interestingly, E. hellem infection desensitized the innate immunity of the host, as shown by cytokine expressions and dendritic cell maturation. We also found that E. hellem infection greatly altered the composition of host gut microbiota. (4) Conclusions: Dex-immunosuppressed mice provide a useful tool for study microsporidiosis and the interactions between microsporidia and host immunity.
Collapse
Affiliation(s)
- Guozhen An
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China; (G.A.); (Y.T.); (B.M.); (M.R.); (Z.Z.)
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China
| | - Yunlin Tang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China; (G.A.); (Y.T.); (B.M.); (M.R.); (Z.Z.)
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China
| | - Biying Mo
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China; (G.A.); (Y.T.); (B.M.); (M.R.); (Z.Z.)
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China
| | - Maoshuang Ran
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China; (G.A.); (Y.T.); (B.M.); (M.R.); (Z.Z.)
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China
| | - Xiao He
- College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China;
| | - Jialing Bao
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China; (G.A.); (Y.T.); (B.M.); (M.R.); (Z.Z.)
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China
| | - Zeyang Zhou
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China; (G.A.); (Y.T.); (B.M.); (M.R.); (Z.Z.)
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China
| |
Collapse
|
21
|
Alauzet C, Cunat L, Wack M, Lanfumey L, Legrand-Frossi C, Lozniewski A, Agrinier N, Cailliez-Grimal C, Frippiat JP. Impact of a Model Used to Simulate Chronic Socio-Environmental Stressors Encountered during Spaceflight on Murine Intestinal Microbiota. Int J Mol Sci 2020; 21:ijms21217863. [PMID: 33114008 PMCID: PMC7672645 DOI: 10.3390/ijms21217863] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/14/2020] [Accepted: 10/21/2020] [Indexed: 12/27/2022] Open
Abstract
During deep-space travels, crewmembers face various physical and psychosocial stressors that could alter gut microbiota composition. Since it is well known that intestinal dysbiosis is involved in the onset or exacerbation of several disorders, the aim of this study was to evaluate changes in intestinal microbiota in a murine model used to mimic chronic psychosocial stressors encountered during a long-term space mission. We demonstrate that 3 weeks of exposure to this model (called CUMS for Chronic Unpredictable Mild Stress) induce significant change in intracaecal β-diversity characterized by an important increase of the Firmicutes/Bacteroidetes ratio. These alterations are associated with a decrease of Porphyromonadaceae, particularly of the genus Barnesiella, a major member of gut microbiota in mice and humans where it is described as having protective properties. These results raise the question of the impact of stress-induced decrease of beneficial taxa, support recent data deduced from in-flight experimentations and other ground-based models, and emphasize the critical need for further studies exploring the impact of spaceflight on intestinal microbiota in order to propose strategies to countermeasure spaceflight-associated dysbiosis and its consequences on health.
Collapse
Affiliation(s)
- Corentine Alauzet
- Stress Immunity Pathogens unit (SIMPA), EA 7300, Université de Lorraine, F-54000 Nancy, France; (L.C.); (C.L.-F.); (A.L.); (C.C.-G.); (J.-P.F.)
- CHRU-Nancy, Service de Microbiologie, F-54000 Nancy, France
- Correspondence: ; Tel.: +33-383-153-938
| | - Lisiane Cunat
- Stress Immunity Pathogens unit (SIMPA), EA 7300, Université de Lorraine, F-54000 Nancy, France; (L.C.); (C.L.-F.); (A.L.); (C.C.-G.); (J.-P.F.)
| | - Maxime Wack
- Département d’Informatique Médicale, Biostatistiques et Santé Publique, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France;
- Centre de Recherche des Cordeliers, INSERM, UMRS 1138, Université de Paris, 75006 Paris, France
| | - Laurence Lanfumey
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Université de Paris, F-75014 Paris, France;
| | - Christine Legrand-Frossi
- Stress Immunity Pathogens unit (SIMPA), EA 7300, Université de Lorraine, F-54000 Nancy, France; (L.C.); (C.L.-F.); (A.L.); (C.C.-G.); (J.-P.F.)
| | - Alain Lozniewski
- Stress Immunity Pathogens unit (SIMPA), EA 7300, Université de Lorraine, F-54000 Nancy, France; (L.C.); (C.L.-F.); (A.L.); (C.C.-G.); (J.-P.F.)
- CHRU-Nancy, Service de Microbiologie, F-54000 Nancy, France
| | - Nelly Agrinier
- CHRU-Nancy, INSERM, Université de Lorraine, CIC, Epidémiologie Clinique, F-54000 Nancy, France;
| | - Catherine Cailliez-Grimal
- Stress Immunity Pathogens unit (SIMPA), EA 7300, Université de Lorraine, F-54000 Nancy, France; (L.C.); (C.L.-F.); (A.L.); (C.C.-G.); (J.-P.F.)
| | - Jean-Pol Frippiat
- Stress Immunity Pathogens unit (SIMPA), EA 7300, Université de Lorraine, F-54000 Nancy, France; (L.C.); (C.L.-F.); (A.L.); (C.C.-G.); (J.-P.F.)
| |
Collapse
|
22
|
Ferreira C, Viana SD, Reis F. Gut Microbiota Dysbiosis-Immune Hyperresponse-Inflammation Triad in Coronavirus Disease 2019 (COVID-19): Impact of Pharmacological and Nutraceutical Approaches. Microorganisms 2020; 8:E1514. [PMID: 33019592 PMCID: PMC7601735 DOI: 10.3390/microorganisms8101514] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 09/29/2020] [Indexed: 12/15/2022] Open
Abstract
Coronavirus Disease 2019 (COVID-19) is a pandemic infection caused by a novel coronavirus named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Patients present a complex clinical picture that, in severe cases, evolves to respiratory, hepatic, gastrointestinal, and neurological complications, and eventually death. The underlying pathophysiological mechanisms are complex and multifactorial and have been summarized as a hyperresponse of the immune system that originates an inflammatory/cytokine storm. In elderly patients, particularly in those with pre-existing cardiovascular, metabolic, renal, and pulmonary disorders, the disease is particularly severe, causing prolonged hospitalization at intensive care units (ICU) and an increased mortality rate. Curiously, the same populations have been described as more prone to a gut microbiota (GM) dysbiosis profile. Intestinal microflora plays a major role in many metabolic and immune functions of the host, including to educate and strengthen the immune system to fight infections, namely of viral origin. Notably, recent studies suggest the existence of GM dysbiosis in COVID-19 patients. This review article highlights the interplay between the triad GM dysbiosis-immune hyperresponse-inflammation in the individual resilience/fragility to SARS-CoV-2 infection and presents the putative impact of pharmacological and nutraceutical approaches on the triumvirate, with focus on GM.
Collapse
Affiliation(s)
- Carolina Ferreira
- Institute of Pharmacology & Experimental Therapeutics, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal;
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-075 Coimbra, Portugal
| | - Sofia D. Viana
- Institute of Pharmacology & Experimental Therapeutics, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal;
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-075 Coimbra, Portugal
- Polytechnic Institute of Coimbra, ESTESC-Coimbra Health School, Pharmacy, 3046-854 Coimbra, Portugal
| | - Flávio Reis
- Institute of Pharmacology & Experimental Therapeutics, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal;
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-075 Coimbra, Portugal
| |
Collapse
|
23
|
Viana SD, Nunes S, Reis F. ACE2 imbalance as a key player for the poor outcomes in COVID-19 patients with age-related comorbidities - Role of gut microbiota dysbiosis. Ageing Res Rev 2020; 62:101123. [PMID: 32683039 PMCID: PMC7365123 DOI: 10.1016/j.arr.2020.101123] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/05/2020] [Accepted: 07/13/2020] [Indexed: 02/06/2023]
Abstract
COVID-19 patients with pre-existing age-related comorbidities have poor outcomes. Gut microbiota dysbiosis is associated with ageing and age-related diseases. Viral-mediated ACE2 shedding favors poor outcomes by RAS-dependent mechanisms. Viral-mediated ACE2 shedding favors poor outcomes by RAS-independent gut dysbiosis. Potential of ACE2 and gut microbiota-based therapeutic opportunities for COVID-19.
Coronavirus disease 19 (COVID-19) is a pandemic condition caused by the new coronavirus SARS-CoV-2. The typical symptoms are fever, cough, shortness of breath, evolving to a clinical picture of pneumonia and, ultimately, death. Nausea and diarrhea are equally frequent, suggesting viral infection or transmission via the gastrointestinal-enteric system. SARS-CoV-2 infects human cells by using angiotensin converting enzyme 2 (ACE2) as a receptor, which is cleaved by transmembrane proteases during host cells infection, thus reducing its activities. ACE2 is a relevant player in the renin-angiotensin system (RAS), counterbalancing the deleterious effects of angiotensin II. Furthermore, intestinal ACE2 functions as a chaperone for the aminoacid transporter B0AT1. It has been suggested that B0AT1/ACE2 complex in the intestinal epithelium regulates gut microbiota (GM) composition and function, with important repercussions on local and systemic immune responses against pathogenic agents, namely virus. Notably, productive infection of SARS-CoV-2 in ACE2+ mature human enterocytes and patients’ GM dysbiosis was recently demonstrated. This review outlines the evidence linking abnormal ACE2 functions with the poor outcomes (higher disease severity and mortality rate) in COVID-19 patients with pre-existing age-related comorbidities and addresses a possible role for GM dysbiosis. The article culminates with the therapeutics opportunities based on these pathways.
Collapse
|