1
|
He L, Li X, Jiang S, Ou Y, Wang S, Shi N, Yang Z, Yuan JL, Silverman G, Niu H. The influence of the gut microbiota on B cells in autoimmune diseases. Mol Med 2025; 31:149. [PMID: 40264032 PMCID: PMC12016346 DOI: 10.1186/s10020-025-01195-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 04/01/2025] [Indexed: 04/24/2025] Open
Abstract
Mounting evidence shows that gut microbiota communities and the human immune system coexist and influence each other, and there are a number of reports of a correlation between specific changes in gut microbiota and the occurrence of autoimmune diseases. B lymphocytes play a central role in the regulation of both gut microbiota communities and in autoimmune diseases. Here, we summarize evidence of the influence of gut microbiota-B cell pathways on autoimmune diseases and how B cells regulate microorganisms, which provides mechanistic insights with relevance for identification of potential therapeutic targets and related fields.
Collapse
Affiliation(s)
- Lun He
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education; Guangzhou Key Laboratory for Germ-free Animals and Microbiota Application, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Xin Li
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education; Guangzhou Key Laboratory for Germ-free Animals and Microbiota Application, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Shan Jiang
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education; Guangzhou Key Laboratory for Germ-free Animals and Microbiota Application, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Yanhua Ou
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education; Guangzhou Key Laboratory for Germ-free Animals and Microbiota Application, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Shanshan Wang
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education; Guangzhou Key Laboratory for Germ-free Animals and Microbiota Application, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Na Shi
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education; Guangzhou Key Laboratory for Germ-free Animals and Microbiota Application, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Zhongshan Yang
- Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, China
| | - Jia-Li Yuan
- Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, China.
| | - Gregg Silverman
- Division of Rheumatology, New York University School of Medicine, New York, NY, 10016, USA.
| | - Haitao Niu
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education; Guangzhou Key Laboratory for Germ-free Animals and Microbiota Application, School of Medicine, Jinan University, Guangzhou, 510632, China.
- Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, China.
| |
Collapse
|
2
|
Pan D, Hao Y, Tao Y, Li B, Cheng L. The influence of microorganisms on bone homeostasis in apical periodontitis. Arch Oral Biol 2025; 170:106153. [PMID: 39644768 DOI: 10.1016/j.archoralbio.2024.106153] [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: 10/23/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/09/2024]
Abstract
OBJECTIVE This review aims to provide an overview of the role of microorganisms in the onset and progression of periapical diseases, particularly regarding their effects on bone homeostasis. DESIGN The search for this narrative review was conducted in PubMed, Web of Science and Google Scholar using relevant keywords, including checking reference lists of journal articles by hand searching. RESULTS Microorganisms directly promote osteoclasts through pathways such as nuclear factor-κB (NF-κB) and extracellular regulated protein kinases (ERK), while inhibiting osteoblasts function by interfering with the wingless-related integration site (Wnt)/β-catenin pathway in the periapical area. Moreover, microorganisms indirectly regulate periapical bone homeostasis by inducing programmed cell death and modulating the immune microenvironment through the activation of innate immunity via pattern-recognition receptors (PRRs) and subsequent cascades of responses. Among these microorganisms, Enterococcus faecalis, Porphyromonas gingivalis and Fusobacterium nucleatum play significant roles. CONCLUSION Microorganisms regulate pathways such as NF-ĸB and Wnt/β-catenin, as well as programmed cell death and the immune microenvironment in the periapical area, thereby disrupting bone homeostasis.
Collapse
Affiliation(s)
- Dan Pan
- West China School of Stomatology (WCSS), Sichuan University, No. 14, Section 3, Renmin Nanlu, Chengdu 610041, China; West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Yu Hao
- West China School of Stomatology (WCSS), Sichuan University, No. 14, Section 3, Renmin Nanlu, Chengdu 610041, China; State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Stomatology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Yuyan Tao
- West China School of Stomatology (WCSS), Sichuan University, No. 14, Section 3, Renmin Nanlu, Chengdu 610041, China; West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Bolei Li
- West China School of Stomatology (WCSS), Sichuan University, No. 14, Section 3, Renmin Nanlu, Chengdu 610041, China; State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Lei Cheng
- West China School of Stomatology (WCSS), Sichuan University, No. 14, Section 3, Renmin Nanlu, Chengdu 610041, China; State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
3
|
Jansen VL, Davids M, van Mourik DJ, Levels JH, Coppens M, Middeldorp S, Nieuwdorp M, van Mens TE. Gut microbiome composition and intestinal immunity in antiphospholipid syndrome patients versus healthy controls. Lupus 2024; 33:1373-1378. [PMID: 39152759 PMCID: PMC11443740 DOI: 10.1177/09612033241274515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/19/2024]
Abstract
INTRODUCTION The gut microbiome is recognized as a factor that could potentially contribute to the persistent antibodies of antiphospholipid syndrome (APS). Gut microbial interventions can both induce and mitigate APS in mice. In human APS patients, anti-beta-2-glycoprotein I (β2GP-1) titers correlate with antibody titers against a gut commensal protein homologous to β2GP-1. AIM To investigate the effect of the intestinal microenvironment on human APS. Methods We cross-sectionally compared intestinal microbiota composition quantified by shotgun sequencing; fecal short chain fatty acids (SCFAs), bacterial metabolites known to affect autoimmune processes; and fecal calprotectin, an intestinal inflammatory marker, in APS patients and healthy controls. RESULTS Neither alpha nor beta diversity of the gut microbiota differed between APS patients (n = 15) and controls (n = 16) and no taxa were differentially abundant. Moreover, fecal SCFAs and fecal calprotectin, did not differ between the groups. CONCLUSION Gut microbiome effects on the APS phenotype are likely not driven by bacterial overabundance, SCFA production or intestinal inflammation.
Collapse
Affiliation(s)
- Valérie Lbi Jansen
- Department of (Experimental) Vascular Medicine, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Pulmonary Hypertension & Thrombosis, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development Research Institute, Amsterdam, The Netherlands
| | - Mark Davids
- Department of (Experimental) Vascular Medicine, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Dagmar Jm van Mourik
- Department of (Experimental) Vascular Medicine, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Pulmonary Hypertension & Thrombosis, Amsterdam, The Netherlands
- Department of Medicine - Thrombosis and Haemostasis, Leiden University Medical Center, Leiden, The Netherlands
| | - Johannes Hm Levels
- Department of (Experimental) Vascular Medicine, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Michiel Coppens
- Department of (Experimental) Vascular Medicine, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Pulmonary Hypertension & Thrombosis, Amsterdam, The Netherlands
| | - Saskia Middeldorp
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Max Nieuwdorp
- Department of (Experimental) Vascular Medicine, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Thijs E van Mens
- Department of (Experimental) Vascular Medicine, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Pulmonary Hypertension & Thrombosis, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development Research Institute, Amsterdam, The Netherlands
- Department of Medicine - Thrombosis and Haemostasis, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
4
|
Fries-Craft K, Schmitz-Esser S, Bobeck EA. Broiler chicken distal jejunum microbial communities are more responsive to coccidiosis or necrotic enteritis challenge than dietary anti-interleukin-10 in a model using Salmonella Typhimurium- Eimeria maxima- Clostridium perfringens coinfection. Poult Sci 2024; 103:104000. [PMID: 39002369 PMCID: PMC11519688 DOI: 10.1016/j.psj.2024.104000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/10/2024] [Accepted: 06/19/2024] [Indexed: 07/15/2024] Open
Abstract
Dietary anti-interleukin (IL)-10 antibodies may protect broiler performance during coccidiosis by inhibiting Eimeria host-evasion pathways; however, anti-IL-10's effects on microbial communities during coccidiosis and secondary Clostridium perfringens (necrotic enteritis) challenge is unknown. The study objectives were to assess the jejunal microbiota of broilers fed anti-IL-10 during E. maxima ± C. perfringens challenge. Two replicate studies using Ross 308 chicks placed in wire-floor cages (32 cages/ replicate study; 20 chicks/ cage) were conducted, with chicks assigned to diets ± 0.03% anti-IL-10 for 25 d. In both replicate studies, challenge-designated chicks were inoculated with 1 × 108Salmonella Typhimurium colony forming units (CFU) at placement. On d14, S. Typhimurium-inoculated chicks were gavaged with 15,000 sporulated Eimeria maxima M6 oocysts and half the E. maxima-challenged chicks received 1×108C. perfringens CFUs on d 18 and 19. Six chicks/ treatment were euthanized for distal jejunum content collection at baseline (d 14), 7 d post-inoculation (pi) with E. maxima/ 3 dpi with C. perfringens (peak) or 11 dpi with E. maxima/ 7 dpi with C. perfringens (post-peak) for 16S rRNA gene amplicon sequencing. Sequences were quality screened (Mothur V.1.43.0) and clustered into de novo operation taxonomical units (OTU; 99% similarity) using the SILVA reference database (v138). Alpha diversity and log-transformed relative abundance data were analyzed in SAS 9.4 with replicate study, diet, challenge, and timepoint main effects plus associated interactions (P ≤ 0.05). Few baseline changes were observed, but E. maxima ± C. perfringens challenge reduced Romboutsia and Staphylococcus relative abundance 4- to 800-fold in both replicate studies (P ≤ 0.008). At peak challenge with secondary C. perfringens, feeding anti-IL-10 instead of the control diet reduced Clostridium sensu stricto 1 relative abundance 13- and 1,848-fold in both replicate studies (P < 0.0001); however, OTUs identified as C. perfringens were not affected by dietary anti-IL-10. These results indicate that anti-IL-10 does not affect the jejunal microbiota of unchallenged broilers, while coccidiosis or necrotic enteritis challenge generally contributed to greater microbiota alterations than diet.
Collapse
Affiliation(s)
- K Fries-Craft
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| | - S Schmitz-Esser
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA; Interdepartmental Graduate Microbiology Program, Iowa State University, Ames, IA 50011, USA
| | - E A Bobeck
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
5
|
Chatterjee S, Leach ST, Lui K, Mishra A. Symbiotic symphony: Understanding host-microbiota dialogues in a spatial context. Semin Cell Dev Biol 2024; 161-162:22-30. [PMID: 38564842 DOI: 10.1016/j.semcdb.2024.03.001] [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: 10/31/2023] [Revised: 02/23/2024] [Accepted: 03/20/2024] [Indexed: 04/04/2024]
Abstract
Modern precision sequencing techniques have established humans as a holobiont that live in symbiosis with the microbiome. Microbes play an active role throughout the life of a human ranging from metabolism and immunity to disease tolerance. Hence, it is of utmost significance to study the eukaryotic host in conjunction with the microbial antigens to obtain a complete picture of the host-microbiome crosstalk. Previous attempts at profiling host-microbiome interactions have been either superficial or been attempted to catalogue eukaryotic transcriptomic profile and microbial communities in isolation. Additionally, the nature of such immune-microbial interactions is not random but spatially organised. Hence, for a holistic clinical understanding of the interplay between hosts and microbiota, it's imperative to concurrently analyze both microbial and host genetic information, ensuring the preservation of their spatial integrity. Capturing these interactions as a snapshot in time at their site of action has the potential to transform our understanding of how microbes impact human health. In examining early-life microbial impacts, the limited presence of communities compels analysis within reduced biomass frameworks. However, with the advent of spatial transcriptomics we can address this challenge and expand our horizons of understanding these interactions in detail. In the long run, simultaneous spatial profiling of host-microbiome dialogues can have enormous clinical implications especially in gaining mechanistic insights into the disease prognosis of localised infections and inflammation. This review addresses the lacunae in host-microbiome research and highlights the importance of profiling them together to map their interactions while preserving their spatial context.
Collapse
Affiliation(s)
- Soumi Chatterjee
- Telethon Kids Institute, Perth Children Hospital, Perth, Western Australia 6009, Australia; Curtin Medical School, Curtin University, Perth, Western Australia 6102, Australia
| | - Steven T Leach
- Discipline Paediatrics, School of Clinical Medicine, University of New South Wales, Sydney 2052, Australia
| | - Kei Lui
- Department of Newborn Care, Royal Hospital for Women and Discipline of Paediatrics and Child Health, School of Clinical Medicine, Faculty of Medicine, University of New South Wales, Sydney 2052, Australia
| | - Archita Mishra
- Telethon Kids Institute, Perth Children Hospital, Perth, Western Australia 6009, Australia; Curtin Medical School, Curtin University, Perth, Western Australia 6102, Australia.
| |
Collapse
|
6
|
Fan J, Kang H, Lv M, Zhai Y, Jia Y, Yang Z, Shi C, Zhou C, Diao L, Li J, Jin X, Liu S, Kristiansen K, Zhang P, Chen J, Li S. Taxonomic composition and functional potentials of gastrointestinal microbiota in 12 wild-stranded cetaceans. Front Microbiol 2024; 15:1394745. [PMID: 39268538 PMCID: PMC11390675 DOI: 10.3389/fmicb.2024.1394745] [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: 03/02/2024] [Accepted: 08/02/2024] [Indexed: 09/15/2024] Open
Abstract
Cetaceans play a crucial role in marine ecosystems; however, research on their gastrointestinal microbiota remains limited due to sampling constraints. In this study, we collected hindgut samples from 12 stranded cetaceans and performed 16S rRNA gene amplicon sequencing to investigate microbial composition and functional potentials. Analysis of ZOTUs profiles revealed that the phyla Firmicutes, Proteobacteria, and Bacteroidetes dominated all hindgut samples. However, unique microbial profiles were observed among different cetacean species, with significant separation of gut microbiota communities according to biological evolutionary lineages. Different genera that contain pathogens were observed distinguishing delphinids from physeteroids/ziphiids. Delphinid samples exhibited higher abundances of Vibrio, Escherichia, and Paeniclostridium, whereas physeteroid and ziphiid samples showed higher abundances of Pseudomonas, Enterococcus, and Intestinimonas. Functional analysis indicated convergence in the gut microbiota among all cetaceans, with shared bacterial infection pathways across hindgut samples. In addition, a comparison of the gastrointestinal microbial composition between a stranded short-finned pilot whale (Globicephala macrorhynchus) and a stranded rough-toothed dolphin (Steno bredanensis) using 16S rRNA gene sequencing revealed distinct microbial community structures and functional capacities. To the best of our knowledge, this study represents the first report on the gastrointestinal microbiota of the pantropical spotted dolphin (Stenella attenuata), Blainville's beaked whale (Mesoplodon densirostris), and rough-toothed dolphin, with various comparisons conducted among different cetacean species. Our findings enhance the understanding of microbial composition and diversity in cetacean gastrointestinal microbiota, providing new insights into co-evolution and complex interactions between cetacean microbes and hosts.
Collapse
Affiliation(s)
- Jie Fan
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI Research, Qingdao, China
| | - Hui Kang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Marine Mammal and Marine Bioacoustics Laboratory, Institute of Deep-sea Science and Engineering, Chinese Academy of Sciences, Sanya, China
| | | | - Yuhuan Zhai
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Marine Mammal and Marine Bioacoustics Laboratory, Institute of Deep-sea Science and Engineering, Chinese Academy of Sciences, Sanya, China
| | | | - Zixin Yang
- Marine Mammal and Marine Bioacoustics Laboratory, Institute of Deep-sea Science and Engineering, Chinese Academy of Sciences, Sanya, China
| | | | | | | | | | - Xiaowei Jin
- China National Environmental Monitoring Centre, Beijing, China
| | | | - Karsten Kristiansen
- Qingdao Key Laboratory of Marine Genomics, and Qingdao-Europe Advanced Institute for Life Sciences, BGI Research, Qingdao, China
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Peijun Zhang
- Marine Mammal and Marine Bioacoustics Laboratory, Institute of Deep-sea Science and Engineering, Chinese Academy of Sciences, Sanya, China
| | - Jianwei Chen
- BGI Research, Qingdao, China
- Qingdao Key Laboratory of Marine Genomics, and Qingdao-Europe Advanced Institute for Life Sciences, BGI Research, Qingdao, China
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Songhai Li
- Marine Mammal and Marine Bioacoustics Laboratory, Institute of Deep-sea Science and Engineering, Chinese Academy of Sciences, Sanya, China
- The Innovation Research Center for Aquatic Mammals, and Key Laboratory of Aquatic Biodiversity and Conservation of the Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
7
|
Arioz Tunc H, Childs CE, Swann JR, Calder PC. The effect of oral probiotics on response to vaccination in older adults: a systematic review of randomised controlled trials. Age Ageing 2024; 53:ii70-ii79. [PMID: 38745493 DOI: 10.1093/ageing/afae033] [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: 12/14/2023] [Indexed: 05/16/2024] Open
Abstract
This systematic review evaluated the impact of oral probiotics on the immune response to vaccination in older people. A literature search was performed in three electronic databases up to January 2023. Randomised controlled trials (RCTs) conducted in older people (age ≥ 60 years) investigating oral probiotics and vaccine response outcomes were included. Characteristics and outcome data of the included studies were extracted and analysed and study quality was assessed using the Cochrane Risk of Bias Tool for randomised trials. Ten RCTs involving 1,560 participants, reported in 9 papers, were included. Nine studies involved the seasonal influenza vaccine and one a COVID-19 vaccine. All studies used lactobacilli, some in combination with bifidobacteria. Studies reported outcomes including anti-vaccine antibody titres or concentrations, seroconversion and seroprotection. When comparing antibody titres, seroprotection rate and seroconversion rate between probiotic and placebo groups expressed as a response ratio, the weighted mean values were 1.29, 1.16 and 2.00, respectively. Meta-analysis showed that probiotics increase seroconversion rates to all three strains of the seasonal influenza vaccine: odds ratio (95% confidence interval) 2.74 (1.31, 5.70; P = 0.007) for the H1N1 strain; 1.90 (1.04, 3.44; P = 0.04) for the H3N2 strain; 1.72 (1.05, 2.80; P = 0.03) for the B strain. There was a low level of heterogeneity in these findings. Several studies were at high risk of bias due to missing outcome data. Lactobacilli may improve the vaccine response, but further research is needed to be more certain of this.
Collapse
Affiliation(s)
- Hediye Arioz Tunc
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Caroline E Childs
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Jonathan R Swann
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton SO16 6YD, UK
| | - Philip C Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton SO16 6YD, UK
| |
Collapse
|
8
|
Joldrichsen MR, Kim E, Steiner HE, Jeong YJ, Premanandan C, Hsueh W, Ziouzenkova O, Cormet-Boyaka E, Boyaka PN. Loss of Paneth cells dysregulates gut ILC subsets and enhances weight gain response to high fat diet in a mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.29.587349. [PMID: 38617293 PMCID: PMC11014498 DOI: 10.1101/2024.03.29.587349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Obesity has been associated with dysbiosis, but innate mechanisms linking intestinal epithelial cell subsets and obesity remain poorly understood. Using mice lacking Paneth cells (Sox9 ΔIEC mice), small intestinal epithelial cells specialized in the production of antimicrobial products and cytokines, we show that dysbiosis alone does not induce obesity or metabolic disorders. Loss of Paneth cells reduced ILC3 and increased ILC2 numbers in the intestinal lamina propria. High-fat diet (HFD) induced higher weight gain and more severe metabolic disorders in Sox9 ΔIEC mice. Further, HFD enhances the number of ILC1 in the intestinal lamina propria of Sox9 ΔIEC mice and increases intestinal permeability and the accumulation of immune cells (inflammatory macrophages and T cells, and B cells) in abdominal fat tissues of obese Sox9 ΔIEC . Transplantation of fecal materials from Sox9 ΔIEC mice in germ-free mice before HFD further confirmed the regulatory role of Paneth cells for gut ILC subsets and the development of obesity.
Collapse
|
9
|
Brown DG, Murphy M, Cadeddu R, Bell R, Weis A, Chiaro T, Klag K, Morgan J, Coon H, Stephens WZ, Bortolato M, Round JL. Colitis reduces active social engagement in mice and is ameliorated by supplementation with human microbiota members. Nat Commun 2024; 15:2769. [PMID: 38553486 PMCID: PMC10980768 DOI: 10.1038/s41467-024-46733-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 03/08/2024] [Indexed: 04/01/2024] Open
Abstract
Multiple neurological disorders are associated with gastrointestinal (GI) symptoms, including autism spectrum disorder (ASD). However, it is unclear whether GI distress itself can modify aspects of behavior. Here, we show that mice that experience repeated colitis have impaired active social engagement, as measured by interactions with a foreign mouse, even though signs of colitis were no longer present. We then tested the hypothesis that individuals with ASD harbor a microbiota that might differentially influence GI health by performing microbiota transplantation studies into male germfree animals, followed by induction of colitis. Animals that harbor a microbiota from ASD individuals have worsened gut phenotypes when compared to animals colonized with microbiotas from familial neurotypical (NT) controls. We identify the enrichment of Blautia species in all familial NT controls and observe an association between elevated abundance of Bacteroides uniformis and reductions in intestinal injury. Oral treatment with either of these microbes reduces colon injury in mice. Finally, provision of a Blautia isolate from a NT control ameliorates gut injury-associated active social engagement in mice. Collectively, our data demonstrate that past intestinal distress is associated with changes in active social behavior in mice that can be ameliorated by supplementation of members of the human microbiota.
Collapse
Affiliation(s)
- D Garrett Brown
- Department of Pathology, University of Utah School of Medicine, Huntsman Cancer Institute, Division of Microbiology and Immunology, Salt Lake City, UT, USA
| | - Michaela Murphy
- Department of Pathology, University of Utah School of Medicine, Huntsman Cancer Institute, Division of Microbiology and Immunology, Salt Lake City, UT, USA
| | - Roberto Cadeddu
- Department of Pharmacology and Toxicology University of Utah College of Pharmacy, Salt Lake City, UT, USA
| | - Rickesha Bell
- Department of Pathology, University of Utah School of Medicine, Huntsman Cancer Institute, Division of Microbiology and Immunology, Salt Lake City, UT, USA
| | - Allison Weis
- Department of Pathology, University of Utah School of Medicine, Huntsman Cancer Institute, Division of Microbiology and Immunology, Salt Lake City, UT, USA
| | - Tyson Chiaro
- Department of Pathology, University of Utah School of Medicine, Huntsman Cancer Institute, Division of Microbiology and Immunology, Salt Lake City, UT, USA
| | - Kendra Klag
- Department of Pathology, University of Utah School of Medicine, Huntsman Cancer Institute, Division of Microbiology and Immunology, Salt Lake City, UT, USA
| | - Jubel Morgan
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Hilary Coon
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - W Zac Stephens
- Department of Pathology, University of Utah School of Medicine, Huntsman Cancer Institute, Division of Microbiology and Immunology, Salt Lake City, UT, USA
| | - Marco Bortolato
- Department of Pharmacology and Toxicology University of Utah College of Pharmacy, Salt Lake City, UT, USA
| | - June L Round
- Department of Pathology, University of Utah School of Medicine, Huntsman Cancer Institute, Division of Microbiology and Immunology, Salt Lake City, UT, USA.
| |
Collapse
|
10
|
Schmiedová L, Černá K, Li T, Těšický M, Kreisinger J, Vinkler M. Bacterial communities along parrot digestive and respiratory tracts: the effects of sample type, species and time. Int Microbiol 2024; 27:127-142. [PMID: 37222909 PMCID: PMC10830831 DOI: 10.1007/s10123-023-00372-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/25/2023]
Abstract
Digestive and respiratory tracts are inhabited by rich bacterial communities that can vary between their different segments. In comparison with other bird taxa with developed caeca, parrots that lack caeca have relatively lower variability in intestinal morphology. Here, based on 16S rRNA metabarcoding, we describe variation in microbiota across different parts of parrot digestive and respiratory tracts both at interspecies and intraspecies levels. In domesticated budgerigar (Melopsittacus undulatus), we describe the bacterial variation across eight selected sections of respiratory and digestive tracts, and three non-destructively collected sample types (faeces, and cloacal and oral swabs). Our results show important microbiota divergence between the upper and lower digestive tract, but similarities between respiratory tract and crop, and also between different intestinal segments. Faecal samples appear to provide a better proxy for intestinal microbiota composition than the cloacal swabs. Oral swabs had a similar bacterial composition as the crop and trachea. For a subset of tissues, we confirmed the same pattern also in six different parrot species. Finally, using the faeces and oral swabs in budgerigars, we revealed high oral, but low faecal microbiota stability during a 3-week period mimicking pre-experiment acclimation. Our findings provide a basis essential for microbiota-related experimental planning and result generalisation in non-poultry birds.
Collapse
Affiliation(s)
- Lucie Schmiedová
- Department of Zoology, Faculty of Science, Charles University, Prague, Czech Republic.
- Institute of Vertebrate Biology, Czech Academy of Sciences, Brno, Czech Republic.
| | - Kateřina Černá
- Department of Zoology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Tao Li
- Department of Zoology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Martin Těšický
- Department of Zoology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Jakub Kreisinger
- Department of Zoology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Michal Vinkler
- Department of Zoology, Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
11
|
Wu Y, Zhang X, Liu X, Li Y, Han D, Pi Y, Whitmore MA, Lu X, Zhang G, Zheng J, Wang J. Strain specificity of lactobacilli with promoted colonization by galactooligosaccharides administration in protecting intestinal barriers during Salmonella infection. J Adv Res 2024; 56:1-14. [PMID: 36894120 PMCID: PMC10834803 DOI: 10.1016/j.jare.2023.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023] Open
Abstract
INTRODUCTION Galactooligosaccharides (GOS) are lactogenic prebiotics that exert health benefits by stimulating the growth of different Lactobacillus strains in the gastrointestinal (GI) tract. OBJECTIVES This study aimed to investigate the mechanism of action of different GOS-enriched lactobacilli in intestinal health. METHODS Piglets and mice were supplemented with GOS to identify specific enrichment of Lactobacillus. The protective effects of individual GOS-enriched lactobacilli were investigated in Salmonella-infected mice. Macrophage depletion and transcriptome analysis were further performed to assess the involvement of macrophages and the underlying mechanisms of individual lactobacilli. An in vitro cell co-culture system was also used to evaluate the anti-adhesive and anti-invasive activities of lactobacilli against Salmonella in epithelial cells. RESULTS GOS markedly increased the relative abundance of three lactobacilli including L. delbrueckii, L. johnsonii, and L. reuteri in both piglets and mice. Supplementation with GOS further alleviated Salmonella infection in mice. L. delbrueckii (ATCC®BAA 365™), but not L. johnsonii or L. reuteri, enhanced propionate production in the intestinal tract and ameliorated Salmonella-induced intestinal inflammation and barrier dysfunction by suppressing the JAK2-STAT3 signaling and M1 macrophage polarization. L. johnsonii (BNCC 186110), on the other hand, inhibited Salmonella adhesion and invasion of epithelial cells through competitive exclusion. However, L. reuteri (BNCC 186135) failed to protect mice against Salmonella infection. CONCLUSION GOS-enriched lactobacilli show a differential role in protecting against Salmonella-induced intestinal barrier dysfunction and inflammation. Our results provide novel insights into the mechanism of action of GOS and individual Lactobacillus strains in the control and prevention of intestinal inflammatory disorders.
Collapse
Affiliation(s)
- Yujun Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xiangyu Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xiaoyi Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yi Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yu Pi
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Melanie A Whitmore
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Xingmiao Lu
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Guolong Zhang
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Jinkai Zheng
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
12
|
Těšický M, Schmiedová L, Krajzingrová T, Samblas MG, Bauerová P, Kreisinger J, Vinkler M. Nearly (?) sterile avian egg in a passerine bird. FEMS Microbiol Ecol 2024; 100:fiad164. [PMID: 38115624 PMCID: PMC10791042 DOI: 10.1093/femsec/fiad164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 12/02/2023] [Accepted: 12/18/2023] [Indexed: 12/21/2023] Open
Abstract
During early ontogeny, microbiome affects development of the gastrointestinal tract, immunity, and survival in vertebrates. Bird eggs are thought to be (1) initially sterile (sterile egg hypothesis) and (2) colonized after oviposition through horizontal trans-shell migration, or (3) initially seeded with bacteria by vertical transfer from mother oviduct. To date, however, little empirical data illuminate the contribution of these mechanisms to gut microbiota formation in avian embryos. We investigated microbiome of the egg content (day 0; E0-egg), embryonic gut at day 13 (E13) and female faeces in a free-living passerine, the great tit (Parus major), using a methodologically advanced procedure combining 16S rRNA gene sequencing and microbe-specific qPCR assays. Our metabarcoding revealed that the avian egg is (nearly) sterile, but acquires a slightly richer microbiome during the embryonic development. Of the three potentially pathogenic bacteria targeted by qPCR, only Dietzia was found in E0-egg (yet also in negative controls), E13 gut and female samples, which might indicate possible vertical transfer. Unlike in poultry, we have shown that major bacterial colonization of the gut in passerines does not occur before hatching. We emphasize that protocols that carefully check for environmental contamination are critical in studies with low-bacterial biomass samples.
Collapse
Affiliation(s)
- Martin Těšický
- Faculty of Science, Department of Zoology, Charles University, Viničná 7, 128 43 Prague, Czech Republic
- Institute of Vertebrate Biology, v.v.i., The Czech Academy of Sciences, Květná 8, Brno 603 65, Czech Republic
- Institute of Paleonatomy, Domestification Research and History of Veterinary Medicine, Ludwig Maxmilian University of Munich, Kaulbachstr. 37 III, 80539 Munich, Germany
| | - Lucie Schmiedová
- Faculty of Science, Department of Zoology, Charles University, Viničná 7, 128 43 Prague, Czech Republic
- Institute of Vertebrate Biology, v.v.i., The Czech Academy of Sciences, Květná 8, Brno 603 65, Czech Republic
| | - Tereza Krajzingrová
- Faculty of Science, Department of Zoology, Charles University, Viničná 7, 128 43 Prague, Czech Republic
| | - Mercedes Gomez Samblas
- Faculty of Science, Department of Zoology, Charles University, Viničná 7, 128 43 Prague, Czech Republic
- Faculty of Science, Department of Parasitology, Campus Universitario de Fuentenueva, University of Granada, Profesor Adolfo Rancano, 18071 Granada, Spain
| | - Petra Bauerová
- Division of Air Quality, Czech Hydrometeorological Institute
, Tušimice Observatory, Tušimice 6, 432 01 Kadaň, Czech Republic
| | - Jakub Kreisinger
- Faculty of Science, Department of Zoology, Charles University, Viničná 7, 128 43 Prague, Czech Republic
| | - Michal Vinkler
- Faculty of Science, Department of Zoology, Charles University, Viničná 7, 128 43 Prague, Czech Republic
| |
Collapse
|
13
|
Abstract
The microbiota is known to influence several facets of mammalian development, digestion and disease. Most studies of the microbiota have focused on the bacterial component, but the importance of commensal fungi in health and disease is becoming increasingly clear. Although fungi account for a smaller proportion of the microbiota than bacteria by number, they are much larger and therefore account for a substantial proportion of the biomass. Moreover, as fungi are eukaryotes, their metabolic pathways are complex and unique. In this Review, we discuss the evidence for involvement of specific members of the mycobiota in intestinal diseases, including inflammatory bowel disease, colorectal cancer and pancreatic cancer. We also highlight the importance of fungal interactions with intestinal bacteria and with the immune system. Although most studies of commensal fungi have focused on their role in disease, we also consider the beneficial effects of fungal colonies in the gut. The evidence highlights potential opportunities to target fungi and their interactions for therapeutic purposes.
Collapse
Affiliation(s)
- Kyla S Ost
- Department of Immunology and Microbiology, University of Colorado Anschutz School of Medicine, Aurora, CO, USA.
| | - June L Round
- Department of Pathology, School of Medicine, University of Utah, Salt Lake City, UT, USA.
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
14
|
Pinnow N, Chibani CM, Güllert S, Weiland-Bräuer N. Microbial community changes correlate with impaired host fitness of Aurelia aurita after environmental challenge. Anim Microbiome 2023; 5:45. [PMID: 37735458 PMCID: PMC10515101 DOI: 10.1186/s42523-023-00266-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 09/10/2023] [Indexed: 09/23/2023] Open
Abstract
Climate change globally endangers certain marine species, but at the same time, such changes may promote species that can tolerate and adapt to varying environmental conditions. Such acclimatization can be accompanied or possibly even be enabled by a host's microbiome; however, few studies have so far directly addressed this process. Here we show that acute, individual rises in seawater temperature and salinity to sub-lethal levels diminished host fitness of the benthic Aurelia aurita polyp, demonstrated by up to 34% reduced survival rate, shrinking of the animals, and almost halted asexual reproduction. Changes in the fitness of the polyps to environmental stressors coincided with microbiome changes, mainly within the phyla Proteobacteria and Bacteroidota. The absence of bacteria amplified these effects, pointing to the benefit of a balanced microbiota to cope with a changing environment. In a future ocean scenario, mimicked by a combined but milder rise of temperature and salinity, the fitness of polyps was severely less impaired, together with condition-specific changes in the microbiome composition. Our results show that the effects on host fitness correlate with the strength of environmental stress, while salt-conveyed thermotolerance might be involved. Further, a specific, balanced microbiome of A. aurita polyps supports the host's acclimatization. Microbiomes may provide a means for acclimatization, and microbiome flexibility can be a fundamental strategy for marine animals to adapt to future ocean scenarios and maintain biodiversity and ecosystem functioning.
Collapse
Affiliation(s)
- Nicole Pinnow
- General Microbiology, Kiel University, Am Botanischen Garten 1-9, 24118, Kiel, Germany
| | - Cynthia M Chibani
- General Microbiology, Kiel University, Am Botanischen Garten 1-9, 24118, Kiel, Germany
| | - Simon Güllert
- General Microbiology, Kiel University, Am Botanischen Garten 1-9, 24118, Kiel, Germany
- Current address: Sysmex Inostics GmbH, Falkenried 88, 20251, Hamburg, Germany
| | - Nancy Weiland-Bräuer
- General Microbiology, Kiel University, Am Botanischen Garten 1-9, 24118, Kiel, Germany.
| |
Collapse
|
15
|
Vorobyev A, Ludwig RJ. Forschung für die Praxis: Ernährung und Mikrobiom bei Autoimmunkrankheiten. J Dtsch Dermatol Ges 2023; 21:958-963. [PMID: 37700405 DOI: 10.1111/ddg.15101_g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 03/17/2023] [Indexed: 09/14/2023]
Abstract
ZusammenfassungDie Häufigkeit von Autoimmunerkrankungen in Industrieländern hat während der letzten Jahrzehnte ständig zugenommen. Diese Erkrankungen führen zu erhöhter Sterblichkeit sowie anhaltender Beeinträchtigung der Lebensqualität der Patienten und bedeuten eine große medizinische Belastung. Die Behandlung von Autoimmunkrankheiten beruht häufig auf unspezifischer Immunsuppression, was das Risiko von Infektionskrankheiten und Krebsmanifestationen erhöht. Die Pathogenese von Autoimmunerkrankungen ist komplex und umfasst nicht nur genetische Faktoren, sondern auch Umwelteinflüsse, die als Grund für die Zunahme von Autoimmunerkrankungen angesehen werden. Zahlreiche Umweltfaktoren wie Infektionen, Rauchen, Medikamente oder Ernährung können das Auftreten von Autoimmunität entweder fördern oder verhindern. Die Mechanismen der Beeinflussung durch Umwelteinflüsse sind jedoch komplex und derzeit noch nicht eindeutig geklärt. Die Entschlüsselung dieser Wechselwirkungen könnte unser Verständnis der Autoimmunität verbessern und neue Behandlungsmöglichkeiten für die Patienten eröffnen.
Collapse
Affiliation(s)
- Artem Vorobyev
- Abteilung für Dermatologie, Allergologie und Venerologie, Universitätsklinikum Schleswig-Holstein, Campus Lübeck
- Lübecker Institut für experimentelle Dermatologie, Lübeck
| | - Ralf J Ludwig
- Abteilung für Dermatologie, Allergologie und Venerologie, Universitätsklinikum Schleswig-Holstein, Campus Lübeck
- Lübecker Institut für experimentelle Dermatologie, Lübeck
| |
Collapse
|
16
|
Vorobyev A, Ludwig RJ. Research in practice: Diet and microbiome in autoimmune diseases. J Dtsch Dermatol Ges 2023; 21:958-962. [PMID: 37235511 DOI: 10.1111/ddg.15101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 03/06/2023] [Accepted: 03/17/2023] [Indexed: 05/28/2023]
Abstract
The incidence of autoimmune diseases in industrialized countries is constantly increasing over past decades. These diseases lead to increased mortality and persistent reduction in quality of life of the patients, posing a severe medical burden. Treatment of autoimmune diseases is often based on unspecific immune suppression, increasing the risk of infectious diseases as well as cancer manifestation. Pathogenesis of autoimmune conditions is complex and includes not only genetic factors, but also environmental influence, which is considered to be the reason for the rise of incidence of autoimmune diseases. Environmental factors comprise numerous elements, such as infections, smoking, medication, diet etc., which can either promote or prevent the onset of autoimmunity. However, the mechanisms of environmental influence are complex and for this moment not clearly understood. Deciphering of these interactions could enhance our comprehension of autoimmunity and provide some novel treatment options for the patients.
Collapse
Affiliation(s)
- Artem Vorobyev
- Department of Dermatology, Allergology and Venereology, University Hospital Schleswig-Holstein, Campus Lübeck, Germany
- Lübeck Institute of Experimental Dermatology, Lübeck, Germany
| | - Ralf J Ludwig
- Department of Dermatology, Allergology and Venereology, University Hospital Schleswig-Holstein, Campus Lübeck, Germany
- Lübeck Institute of Experimental Dermatology, Lübeck, Germany
| |
Collapse
|
17
|
Song Y, Li X, Huang D, Song H. Macrophages in periapical lesions: Potential roles and future directions. Front Immunol 2022; 13:949102. [PMID: 36131939 PMCID: PMC9483141 DOI: 10.3389/fimmu.2022.949102] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
Periapical lesions are infectious diseases that occur in the apical region of teeth. They result in the destruction of alveolar bone and are usually accompanied by swelling, pain, and possible systemic impacts. A complex interaction between pathogens and the host immune system determines the development, progression, and outcome of periapical lesions. The lesions, if not treated promptly, may cause resorption of bone tissue, destruction of the periodontal ligament, and loss of the affected teeth, all of which can severely worsen the quality of life of patients, often at considerable economic cost to both patients and medical organizations. Macrophages are a group of heterogeneous cells that have many roles in the development of infections, destruction and reconstruction of bone tissues, and microbe–host interactions. However, the differential and comprehensive polarization of macrophages complicates the understanding of the regulatory mechanism of periapical lesion progression. This report provides a comprehensive review of recent advances in our knowledge of the potential role of macrophages in determining the turnover of human periapical lesions. For example, macrophage differentiation might indicate whether the lesions are stable or progressing while the extent of bacteria invasion could regulate the differentiation and function of macrophages involved in the periapical lesion. In addition, alternative strategies for the treatment of apical periodontitis are discussed.
Collapse
Affiliation(s)
- Yao Song
- Department of Stomatology, Chengdu Second People’s Hospital, Chengdu, China
| | - Xinying Li
- Department of Stomatology, Chengdu Second People’s Hospital, Chengdu, China
| | - Dingming Huang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Hongjie Song, ; Dingming Huang,
| | - Hongjie Song
- Department of Stomatology, Chengdu Second People’s Hospital, Chengdu, China
- *Correspondence: Hongjie Song, ; Dingming Huang,
| |
Collapse
|
18
|
Cancer immunotherapy resistance: The impact of microbiome-derived short-chain fatty acids and other emerging metabolites. Life Sci 2022; 300:120573. [DOI: 10.1016/j.lfs.2022.120573] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 04/10/2022] [Accepted: 04/18/2022] [Indexed: 12/12/2022]
|
19
|
Killian M, van Mens TE. Risk of Thrombosis, Pregnancy Morbidity or Death in Antiphospholipid Syndrome. Front Cardiovasc Med 2022; 9:852777. [PMID: 35299976 PMCID: PMC8921454 DOI: 10.3389/fcvm.2022.852777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/07/2022] [Indexed: 12/12/2022] Open
Abstract
The antiphospholipid syndrome is an autoimmune disease characterized by thrombosis and pregnancy morbidity. The manifestations are caused by antibodies targeting cell membrane phospholipids and/or associated proteins. The triggers leading to these antibodies' production are unknown but recent work suggests cross-reactivity between the autoantigens and peptides produced by the intestinal microbiome. Work on how the autoantibodies could cause clinical manifestations implicates different mechanisms. Binding to surface proteins of different cell types can induce intracellular signaling leading to cell activation and tissue factor expression. Complement activation and neutrophil extracellular-traps are also involved, and recent evidence implicates endothelial protein C receptor-lysobisphosphatidic acid complex. Pregnancy is a high-risk situation for antiphospholipid syndrome patients due to the increased risk of thrombosis and obstetric complications. Epidemiological and clinical research on APS is hampered by heterogeneity in populations, testing and treatment strategies. About one in 10 to one in fifty APS pregnancies is complicated by thrombosis, despite treatment. Pregnant patients with prior thrombosis are prescribed therapeutic dose heparins and low dose aspirin. Without prior thrombosis a prophylactic dose is used. The most frequent obstetrical manifestation is recurrent early pregnancy loss. The association of APS antibodies with late pregnancy loss is stronger, however. Prevention of recurrence is achieved with aspirin and prophylactic dose heparin, although the evidence is of low certainty. The third obstetrical classifying manifestation comprises preterm delivery due to placenta-mediated complications and is treated in subsequent pregnancies with aspirin with or without prophylactic dose heparin, again based on low quality evidence. New therapies are under investigation.
Collapse
Affiliation(s)
- Martin Killian
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, Saint-Étienne, France.,Internal Medicine Department, Saint-Etienne University Hospital, Saint-Étienne, France
| | - Thijs E van Mens
- Amsterdam UMC, Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam Reproduction and Development, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
20
|
Zhi L, Ai D, Yong M, Bao H, Han B, Sun B, Tu Y, Er D. The effects of genital myiasis on the diversity of the vaginal microbiota in female Bactrian camels. BMC Vet Res 2022; 18:87. [PMID: 35248026 PMCID: PMC8897907 DOI: 10.1186/s12917-022-03189-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 02/23/2022] [Indexed: 12/31/2022] Open
Abstract
Background Genital myasis is one of the most important diseases that affects the reproductive organs of Bactrian camels in which can cause serious mechanical damage to the vaginal tissue. The accumulation of bacteria in the vagina of female camels can affect their health and reproductive ability. The effect of this damage is commonly manifested in the vaginal flora and vaginal mucosal immune system. Therefore, this investigation is a study of the diversity of the vaginal flora and the differences between healthy Bactrian camels and those suffering from genital myiasis. Results Vaginal microbiota samples were collected from two groups of female Bactrian camels of the same age. An Illumina MiSeq was used to sequence the 16S rRNA V3-V4 hypervariable sequence in the samples. The results showed that the vaginal microflora of the infected camels had a significantly greater operational taxonomic unit (OTU) value. According to the assessment of the alpha diversity index and the vaginal pH, the diversity index of the infected camel flora was higher than that of the normal camel flora, and the vaginal pH was lower than that of the normal camels (p < 0.01). There were no significant differences between the two groups in the abundance of dominant genera in the Bactrian camel vagina (P > 0.05), indicating that the certain stability is maintained. Conclusions Overall, this comparison revealed the differences and similarities between the vaginal microbiota of Bactrian camels in various health statues. In addition, these data provide a reference point for understanding the types of bacteria that cause genital myiasis affecting the healthy development of Bactrian camels. Supplementary Information The online version contains supplementary material available at 10.1186/s12917-022-03189-5.
Collapse
|
21
|
Ma Y, Guo R, Sun Y, Li X, He L, Li Z, Silverman GJ, Chen G, Gao F, Yuan J, Wei Q, Li M, Lu L, Niu H. Lupus gut microbiota transplants cause autoimmunity and inflammation. Clin Immunol 2021; 233:108892. [PMID: 34813937 DOI: 10.1016/j.clim.2021.108892] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 11/17/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND The etiology of systemic lupus erythematosus (SLE) is multifactorial. Recently, growing evidence suggests that the microbiota plays a role in SLE, yet whether gut microbiota participates in the development of SLE remains largely unknown. To investigate this issue, we carried out 16 s rDNA sequencing analyses in a cohort of 18 female un-treated active SLE patients and 7 female healthy controls, and performed fecal microbiota transplantation from patients and healthy controls to germ-free (GF) mice. RESULTS Compared to the healthy controls, we found no significant different microbial diversity but some significantly different species in SLE patients including Turicibacter genus and other 5 species. Fecal transfer from SLE patients to GF mice caused GF mice to develop a series of lupus-like phenotypic features, including increased serum autoimmune antibodies, imbalanced cytokines, altered distribution of immune cells in mucosal and peripheral immune response, and upregulated expression of genes related to SLE in recipient mice that received SLE fecal microbiota transplantation (FMT). Moreover, the metabolism of histidine was significantly altered in GF mice treated with SLE patient feces, as compared to those which received healthy fecal transplants. CONCLUSIONS Overall, our results describe a causal role of aberrant gut microbiota in contributing to the pathogenesis of SLE. The interplay of gut microbial and histidine metabolism may be one of the mechanisms intertwined with autoimmune activation in SLE.
Collapse
Affiliation(s)
- Yiyangzi Ma
- School of Medicine and Institute of Laboratory Animal Sciences, Jinan University; Guangzhou Key Laboratory of Germ-free Animals and Microbiota Application, Guangzhou 510632, China; Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200001, China
| | - Ruru Guo
- Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200001, China
| | - Yiduo Sun
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing 100730, China; Department of Rheumatology,The First Affiliated Hospital, Zhejiang University School of Medicine (FAHZU), Hangzhou 310003, China
| | - Xin Li
- School of Medicine and Institute of Laboratory Animal Sciences, Jinan University; Guangzhou Key Laboratory of Germ-free Animals and Microbiota Application, Guangzhou 510632, China
| | - Lun He
- School of Medicine and Institute of Laboratory Animal Sciences, Jinan University; Guangzhou Key Laboratory of Germ-free Animals and Microbiota Application, Guangzhou 510632, China
| | - Zhao Li
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing 100730, China; Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| | - Gregg J Silverman
- Division of Rheumatology, New York University School of Medicine, New York, NY 10016, USA
| | - Guobing Chen
- School of Medicine and Institute of Laboratory Animal Sciences, Jinan University; Guangzhou Key Laboratory of Germ-free Animals and Microbiota Application, Guangzhou 510632, China
| | - Feng Gao
- School of Medicine and Institute of Laboratory Animal Sciences, Jinan University; Guangzhou Key Laboratory of Germ-free Animals and Microbiota Application, Guangzhou 510632, China
| | - Jiali Yuan
- School of Basic Medicine, Yunnan University of Traditional Chinese Medicine, China
| | - Qiang Wei
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, China
| | - Mengtao Li
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing 100730, China.
| | - Liangjing Lu
- Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200001, China.
| | - Haitao Niu
- School of Medicine and Institute of Laboratory Animal Sciences, Jinan University; Guangzhou Key Laboratory of Germ-free Animals and Microbiota Application, Guangzhou 510632, China; Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200001, China.
| |
Collapse
|
22
|
Grieves LA, Gloor GB, Bernards MA, MacDougall-Shackleton EA. Preen gland microbiota covary with major histocompatibility complex genotype in a songbird. ROYAL SOCIETY OPEN SCIENCE 2021; 8:210936. [PMID: 34754501 PMCID: PMC8493191 DOI: 10.1098/rsos.210936] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 09/13/2021] [Indexed: 05/30/2023]
Abstract
Pathogen-mediated selection at the major histocompatibility complex (MHC) is thought to promote MHC-based mate choice in vertebrates. Mounting evidence implicates odour in conveying MHC genotype, but the underlying mechanisms remain uncertain. MHC effects on odour may be mediated by odour-producing symbiotic microbes whose community structure is shaped by MHC genotype. In birds, preen oil is a primary source of body odour and similarity at MHC predicts similarity in preen oil composition. Hypothesizing that this relationship is mediated by symbiotic microbes, we characterized MHC genotype, preen gland microbial communities and preen oil chemistry of song sparrows (Melospiza melodia). Consistent with the microbial mediation hypothesis, pairwise similarity at MHC predicted similarity in preen gland microbiota. Counter to this hypothesis, overall microbial similarity did not predict chemical similarity of preen oil. However, permutation testing identified a maximally predictive set of microbial taxa that best reflect MHC genotype, and another set of taxa that best predict preen oil chemical composition. The relative strengths of relationships between MHC and microbes, microbes and preen oil, and MHC and preen oil suggest that MHC may affect host odour both directly and indirectly. Thus, birds may assess MHC genotypes based on both host-associated and microbially mediated odours.
Collapse
Affiliation(s)
- L. A. Grieves
- Department of Biology, University of Western Ontario, London, ON, Canada N6A 5B7
| | - G. B. Gloor
- Department of Biochemistry, University of Western Ontario, London, ON, Canada N6A 5C1
| | - M. A. Bernards
- Department of Biology, University of Western Ontario, London, ON, Canada N6A 5B7
| | | |
Collapse
|
23
|
Li B, Gong T, Hao Y, Zhou X, Cheng L. Mining the Gut Microbiota for Microbial-Based Therapeutic Strategies in Cancer Immunotherapy. Front Oncol 2021; 11:721249. [PMID: 34589427 PMCID: PMC8473692 DOI: 10.3389/fonc.2021.721249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 08/23/2021] [Indexed: 02/05/2023] Open
Abstract
The past two decades witnessed a revolution in our understanding of host–microbiota interactions that led to the concept of the super-organism consisting of a eukaryotic part and a prokaryotic part. Owing to the critical role of gut microbiota in modulating the host immune system, it is not beyond all expectations that more and more evidence indicated that the shift of gut microbiota influenced responses to numerous forms of cancer immunotherapy. Therapy targeting gut microbiota is becoming a promising strategy to improve cancer immunotherapy. In this review, we discuss the role of the gut microbiota in response to cancer immunotherapy, the mechanisms that the gut microbiota influences cancer immunotherapy, and therapeutic strategies targeting gut microbiota to improve cancer immunotherapy.
Collapse
Affiliation(s)
- Bolei Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China.,Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Tao Gong
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Yu Hao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China.,Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China.,Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China.,Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
24
|
Glowacki RWP, Engelhart MJ, Ahern PP. Controlled Complexity: Optimized Systems to Study the Role of the Gut Microbiome in Host Physiology. Front Microbiol 2021; 12:735562. [PMID: 34646255 PMCID: PMC8503645 DOI: 10.3389/fmicb.2021.735562] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/24/2021] [Indexed: 12/26/2022] Open
Abstract
The profound impact of the gut microbiome on host health has led to a revolution in biomedical research, motivating researchers from disparate fields to define the specific molecular mechanisms that mediate host-beneficial effects. The advent of genomic technologies allied to the use of model microbiomes in gnotobiotic mouse models has transformed our understanding of intestinal microbial ecology and the impact of the microbiome on the host. However, despite incredible advances, our understanding of the host-microbiome dialogue that shapes host physiology is still in its infancy. Progress has been limited by challenges associated with developing model systems that are both tractable enough to provide key mechanistic insights while also reflecting the enormous complexity of the gut ecosystem. Simplified model microbiomes have facilitated detailed interrogation of transcriptional and metabolic functions of the microbiome but do not recapitulate the interactions seen in complex communities. Conversely, intact complex communities from mice or humans provide a more physiologically relevant community type, but can limit our ability to uncover high-resolution insights into microbiome function. Moreover, complex microbiomes from lab-derived mice or humans often do not readily imprint human-like phenotypes. Therefore, improved model microbiomes that are highly defined and tractable, but that more accurately recapitulate human microbiome-induced phenotypic variation are required to improve understanding of fundamental processes governing host-microbiome mutualism. This improved understanding will enhance the translational relevance of studies that address how the microbiome promotes host health and influences disease states. Microbial exposures in wild mice, both symbiotic and infectious in nature, have recently been established to more readily recapitulate human-like phenotypes. The development of synthetic model communities from such "wild mice" therefore represents an attractive strategy to overcome the limitations of current approaches. Advances in microbial culturing approaches that allow for the generation of large and diverse libraries of isolates, coupled to ever more affordable large-scale genomic sequencing, mean that we are now ideally positioned to develop such systems. Furthermore, the development of sophisticated in vitro systems is allowing for detailed insights into host-microbiome interactions to be obtained. Here we discuss the need to leverage such approaches and highlight key challenges that remain to be addressed.
Collapse
Affiliation(s)
- Robert W. P. Glowacki
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Morgan J. Engelhart
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Philip P. Ahern
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
25
|
Gogokhia L, Round JL. Immune-bacteriophage interactions in inflammatory bowel diseases. Curr Opin Virol 2021; 49:30-35. [PMID: 34029992 DOI: 10.1016/j.coviro.2021.04.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/25/2021] [Accepted: 04/26/2021] [Indexed: 01/03/2023]
Abstract
Inflammatory bowel disease (IBD), including ulcerative colitis (UC) and Crohn's disease (CD), are influenced by the bacterial and fungal organisms found within the intestine. However, the intestine is also home to a vast number of viral particles, with most of them being viruses that infect prokaryotes, called bacteriophages. While use of bacteriophages to specifically target pathogenic bacterial species involved in IBD is currently under investigation, recent studies have also highlighted that these viral particles can impact the mammalian immune system. IBD is a chronic multi-factorial inflammatory condition with unknown etiology. This review will highlight the current investigations that have revealed that bacteriophage-mammalian immune cell interactions can influence disease processes beyond their known role for infecting bacteria, which might identify novel ways to treat or diagnose IBD.
Collapse
Affiliation(s)
- Lasha Gogokhia
- Department of Pathology, University of Utah School of Medicine, Division of Microbiology and Immunology, UT 84112, United States; Department of Internal Medicine, St. Mary's Hospital, Waterbury, CT 06706, United States
| | - June L Round
- Department of Pathology, University of Utah School of Medicine, Division of Microbiology and Immunology, UT 84112, United States.
| |
Collapse
|
26
|
Affiliation(s)
- Tyson Chiaro
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - June L Round
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Salt Lake City, UT, USA. .,Huntsman Cancer Institute, University of Utah, Circle of Hope Drive, Salt Lake City, UT, USA
| |
Collapse
|
27
|
Wang Y, Nan X, Zhao Y, Jiang L, Wang M, Wang H, Zhang F, Xue F, Hua D, Liu J, Yao J, Xiong B. Rumen microbiome structure and metabolites activity in dairy cows with clinical and subclinical mastitis. J Anim Sci Biotechnol 2021; 12:36. [PMID: 33557959 PMCID: PMC7869221 DOI: 10.1186/s40104-020-00543-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 12/28/2020] [Indexed: 12/17/2022] Open
Abstract
Background Due to the high prevalence and complex etiology, bovine mastitis (BM) is one of the most important diseases to compromise dairy cow health and milk quality. The shift in milk compositions has been widely investigated during mastitis, but recent studies suggested that gastrointestinal microorganism also has a crucial effect on the inflammation of other peripheral tissues and organs, including the mammary gland. However, research focused on the variation of rumen inner-environment during mastitis is still limited. Therefore, the ruminal microbial profiles, metabolites, and milk compositions in cows with different udder health conditions were compared in the present study. Furthermore, the correlations between udder health status and ruminal conditions were investigated. Based on the somatic cell counts (SCC), California mastitis test (CMT) parameters and clinical symptoms of mastitis, 60 lactating Holstein dairy cows with similar body conditions (excepted for the udder health condition) were randomly divided into 3 groups (n = 20 per group) including the healthy (H) group, the subclinical mastitis (SM) group and the clinical mastitis (CM) group. Lactation performance and rumen fermentation parameters were recorded. And rumen microbiota and metabolites were also analyzed via 16S rRNA amplicon sequencing and untargeted metabolomics, respectively. Results As the degree of mastitis increased, rumen lactic acid (LA) (P < 0.01), acetate, propionate, butyrate, valerate (P < 0.001), and total volatile fatty acids (TVFAs) (P < 0.01) concentrations were significantly decreased. In the rumen of CM cows, the significantly increased bacteria related to intestinal and oral inflammation, such as Lachnospiraceae (FDR-adjusted P = 0.039), Moraxella (FDR-adjusted P = 0.011) and Neisseriaceae (FDR-adjusted P = 0.036), etc., were accompanied by a significant increase in 12-oxo-20-dihydroxy-leukotriene B4 (FDR-adjusted P = 5.97 × 10− 9) and 10beta-hydroxy-6beta-isobutyrylfuranoeremophilane (FDR-adjusted P = 3.88 × 10− 10). Meanwhile, in the rumen of SM cows, the Ruminiclostridium_9 (FDR-adjusted P = 0.042) and Enterorhabdus (FDR-adjusted P = 0.043) were increased along with increasing methenamine (FDR-adjusted P = 6.95 × 10− 6), 5-hydroxymethyl-2-furancarboxaldehyde (5-HMF) (FDR-adjusted P = 2.02 × 10− 6) and 6-methoxymellein (FDR-adjusted P = 2.57 × 10− 5). The short-chain fatty acids (SCFAs)-producing bacteria and probiotics in rumen, including Prevoterotoella_1 (FDR-adjusted P = 0.045) and Bifidobacterium (FDR-adjusted P = 0.035), etc., were significantly reduced, with decreasing 2-phenylbutyric acid (2-PBA) (FDR-adjusted P = 4.37 × 10− 6). Conclusion The results indicated that there was a significant shift in the ruminal microflora and metabolites associated with inflammation and immune responses during CM. Moreover, in the rumen of cows affected by SM, the relative abundance of several opportunistic pathogens and the level of metabolites which could produce antibacterial compounds or had a competitive inhibitory effect were all increased. Supplementary Information The online version contains supplementary material available at 10.1186/s40104-020-00543-1.
Collapse
Affiliation(s)
- Yue Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.,College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Xuemei Nan
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Yiguang Zhao
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Linshu Jiang
- Beijing Key Laboratory for Dairy Cow Nutrition, Beijing University of Agriculture, Beijing, 102206, China.
| | - Mengling Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Hui Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Fan Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Fuguang Xue
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.,Engineering Research Center of Feed Development, Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Dengke Hua
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Jun Liu
- Langfang Academy of Agriculture and Forestry, Langfang, 065000, China
| | - Junhu Yao
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Benhai Xiong
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| |
Collapse
|
28
|
Alpízar-Rodríguez D, Finckh A, Gilbert B. The Role of Nutritional Factors and Intestinal Microbiota in Rheumatoid Arthritis Development. Nutrients 2020; 13:nu13010096. [PMID: 33396685 PMCID: PMC7823566 DOI: 10.3390/nu13010096] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 02/07/2023] Open
Abstract
Evidence about the role of nutritional factors and microbiota in autoimmune diseases, and in rheumatoid arthritis (RA) in particular, has grown in recent years, however many controversies remain. The aim of this review is to summarize the role of nutrition and of the intestinal microbiota in the development of RA. We will focus on selected dietary patterns, individual foods and beverages that have been most consistently associated with RA or with the occurrence of systemic autoimmunity associated with RA. We will also review the evidence for a role of the intestinal microbiota in RA development. We propose that diet and digestive microbiota should be considered together in research, as they interact and may both be the target for future preventive interventions in RA.
Collapse
Affiliation(s)
- Deshiré Alpízar-Rodríguez
- Research Unit, Colegio Mexicano de Reumatología, Mexico City 04318, Mexico
- Correspondence: ; Tel.: +52-55-2525-1853
| | - Axel Finckh
- Department of Rheumatology, Geneva University Hospitals, 1206 Geneva, Switzerland; (A.F.); (B.G.)
| | - Benoît Gilbert
- Department of Rheumatology, Geneva University Hospitals, 1206 Geneva, Switzerland; (A.F.); (B.G.)
| |
Collapse
|
29
|
Wood TE, Aksoy E, Hachani A. From Welfare to Warfare: The Arbitration of Host-Microbiota Interplay by the Type VI Secretion System. Front Cell Infect Microbiol 2020; 10:587948. [PMID: 33194832 PMCID: PMC7604300 DOI: 10.3389/fcimb.2020.587948] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/22/2020] [Indexed: 12/17/2022] Open
Abstract
The health of mammals depends on a complex interplay with their microbial ecosystems. Compartments exposed to external environments such as the mucosal surfaces of the gastrointestinal tract accommodate the gut microbiota, composed by a wide range of bacteria. The gut microbiome confers benefits to the host, including expansion of metabolic potential and the development of an immune system that can robustly protect from external and internal insults. The cooperation between gut microbiome and host is enabled in part by the formation of partitioned niches that harbor diverse bacterial phyla. Bacterial secretion systems are commonly employed to manipulate the composition of these local environments. Here, we explore the roles of the bacterial type VI secretion system (T6SS), present in ~25% of gram-negative bacteria, including many symbionts, in the establishment and perturbation of bacterial commensalism, and symbiosis in host mucosal sites. This versatile apparatus drives bacterial competition, although in some cases can also interfere directly with host cells and facilitate nutrient acquisition. In addition, some bacterial pathogens cause disease when their T6SS leads to dysbiosis and subverts host immune responses in defined animal models. This review explores our knowledge of the T6SS in the context of the “host-microbiota-pathogen” triumvirate and examines contexts in which the importance of this secretion system may be underappreciated.
Collapse
Affiliation(s)
- Thomas E Wood
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, United States.,Department of Microbiology, Harvard Medical School, Boston, MA, United States
| | - Ezra Aksoy
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Abderrahman Hachani
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| |
Collapse
|
30
|
Ren L, Zhang J, Zhang T. Immunomodulatory activities of polysaccharides from Ganoderma on immune effector cells. Food Chem 2020; 340:127933. [PMID: 32882476 DOI: 10.1016/j.foodchem.2020.127933] [Citation(s) in RCA: 202] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 07/10/2020] [Accepted: 08/23/2020] [Indexed: 02/04/2023]
Abstract
Polysaccharides are the most abundant bioactive compounds in Ganoderma and have been widely used as dietary supplements in traditional Chinese medicine for thousands of years. Polysaccharides from Ganoderma exhibit unique biological properties, including anti-tumor, anti-inflammatory, and immunomodulatory activities. Herein, the sources and structures of polysaccharides from Ganoderma were presented. This work also reviews the immunomodulatory activities and possible mechanisms of polysaccharides from Ganoderma on different immune effector cells, including lymphocytes and myeloid cells. As an available adjunctive remedy, polysaccharides from Ganoderma can potentially be applied for the modulation of the host immune system, namely the innate immunity, the cellular immunity, and the humoral immunity.
Collapse
Affiliation(s)
- Li Ren
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Jie Zhang
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | - Tiehua Zhang
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW With the emergence of the microbiota as a potential driver of host inflammation, the role of iIgA is becoming increasingly important. This review discusses the current evidence regarding the effects of clinical IgA deficiency on the microbiota, and the possible role of microbial dysbiosis in driving inflammation in PID patients. RECENT FINDINGS The gut microbiota has been investigated in selective IgA deficiency and common variable immunodeficiency, revealing an important role for IgA in maintaining gut microbiota homeostasis, with disparate effects of IgA on symbionts and pathobionts. Although IgA deficiency is associated with microbial translocation and systemic inflammation, this may be partially compensated by adequate IgG and IgM induction in IgA deficiency but not in common variable immunodeficiency. Therapeutic strategies aimed at correction of the microbiota mostly focus on fecal microbiota transplantation. Whether this may reduce systemic inflammation in PID is currently unknown. SUMMARY Clinical IgA deficiency is associated with microbial dysbiosis and systemic inflammation. The evidence for microbiota-targeted therapies in PID is scarce, but indicates that IgA-based therapies may be beneficial, and that fecal microbiota transplantation is well tolerated in patients with antibody deficiency.
Collapse
|
32
|
Choi SC, Brown J, Gong M, Ge Y, Zadeh M, Li W, Croker BP, Michailidis G, Garrett TJ, Mohamadzadeh M, Morel L. Gut microbiota dysbiosis and altered tryptophan catabolism contribute to autoimmunity in lupus-susceptible mice. Sci Transl Med 2020; 12:eaax2220. [PMID: 32641487 PMCID: PMC7739186 DOI: 10.1126/scitranslmed.aax2220] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 10/04/2019] [Accepted: 01/27/2020] [Indexed: 12/12/2022]
Abstract
The autoimmune disease systemic lupus erythematosus (SLE) is characterized by the production of pathogenic autoantibodies. It has been postulated that gut microbial dysbiosis may be one of the mechanisms involved in SLE pathogenesis. Here, we demonstrate that the dysbiotic gut microbiota of triple congenic (TC) lupus-prone mice (B6.Sle1.Sle2.Sle3) stimulated the production of autoantibodies and activated immune cells when transferred into germfree congenic C57BL/6 (B6) mice. Fecal transfer to B6 mice induced autoimmune phenotypes only when the TC donor mice exhibited autoimmunity. Autoimmune pathogenesis was mitigated by horizontal transfer of the gut microbiota between co-housed lupus-prone TC mice and control congenic B6 mice. Metabolomic screening identified an altered distribution of tryptophan metabolites in the feces of TC mice including an increase in kynurenine, which was alleviated after antibiotic treatment. Low dietary tryptophan prevented autoimmune pathology in TC mice, whereas high dietary tryptophan exacerbated disease. Reducing dietary tryptophan altered gut microbial taxa in both lupus-prone TC mice and control B6 mice. Consequently, fecal transfer from TC mice fed a high tryptophan diet, but not a low tryptophan diet, induced autoimmune phenotypes in germfree B6 mice. The interplay of gut microbial dysbiosis, tryptophan metabolism and host genetic susceptibility in lupus-prone mice suggest that aberrant tryptophan metabolism may contribute to autoimmune activation in this disease.
Collapse
Affiliation(s)
- Seung-Chul Choi
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Josephine Brown
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Minghao Gong
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL 32610, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Yong Ge
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL 32610, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Mojgan Zadeh
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL 32610, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Wei Li
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Byron P Croker
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - George Michailidis
- Department of Statistics and the Informatics Institute, University of Florida, Gainesville, FL 32610, USA
| | - Timothy J Garrett
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Mansour Mohamadzadeh
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL 32610, USA.
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Laurence Morel
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
33
|
Bukina Y, Thyhonovska M, Koval M, Marushchak M, Krynytska I, Kamyshnyi A. The effect of immunoregulatory bacteria on the transcriptional activity of Foxp3 and RORyt genes in the gut-associated lymphoid tissue with Salmonella-induced inflammation in the presence of vancomycin and Bacteroides fragilis. IRANIAN JOURNAL OF MICROBIOLOGY 2020. [PMID: 32685120 DOI: 10.18502/ijm.v12i3.3241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Background and Objectives Intestinal microbiota is involved in the development and maintenance of immune homeostasis. This study was conducted to investigate the levels of key immunoregulatory bacteria in the intestinal wall-associated microflora and its effect on the transcriptional activity of the Foxp3 and RORyt genes in the gut-associated lymphoid tissue (GALT) of rats with Salmonella-induced inflammation, both untreated and treated with vancomycin and Bacteroides fragilis. Materials and Methods To determine the levels of immunoregulatory bacteria in GALT of rats Q-PCR was used to identify them by species-specific 16S rDNA genes. Transcriptional activity of Foxp3 and RORyt genes was determined using Q-PCR with reverse transcription. Results In animals treated with both vancomycin and Salmonella, the levels of segmented filamentous bacteria (SFB) increased while Akkermansia muciniphila and Faecalibacterium prausnitzii decreased. In rats that received pretreatment with vancomycin and then were infected with S. Enteritidis and S. Typhimurium, the levels of SFB increased, and the number of Bacteroides-Prevotela group, A. muciniphila, Clostridium spp. clusters XIV, IV, and F. prausnitzii significantly decreased, decreasing Foxp3 and increasing Rorγt mRNA expression. Administration of B. fragilis to animals treated with S. Enteritidis or S. Typhimurium and pre-treated with vancomycin caused a decrease in SFB and Rorγt mRNA levels and conversely, increased the numbers of the Bacteroides-Prevotela group, Clostridium spp. clusters XIV, IV, A. muciniphila, F. prausnitzii and Foxp3 gene expression in GALT. Conclusion Our results suggest that the commensal microorganism B. fragilis may provide a protective role against the development of experimental colitis, which has to be taken into consideration for further clarification of the effective therapeutic strategy of inflammatory bowel diseases, irritable bowel syndrome and necrotising colitis.
Collapse
Affiliation(s)
- Yuliia Bukina
- Department of Microbiology, Virology and Immunology, Zaporizhzhya State Medical University, Zaporizhzhya, Ukraine
| | - Marina Thyhonovska
- Department of Normal Physiology, Zaporizhzhya State Medical University, Zaporizhzhya, Ukraine
| | - Mariya Koval
- Department of General Chemistry, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Mariya Marushchak
- Department of Functional and Laboratory Diagnostics, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Inna Krynytska
- Department of Functional and Laboratory Diagnostics, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Aleksandr Kamyshnyi
- Department of Microbiology, Virology and Immunology, Zaporizhzhya State Medical University, Zaporizhzhya, Ukraine
| |
Collapse
|
34
|
Wu M, Wang F, Yang J, Li P, Yan D, Yang Y, Zhang W, Ren J, Zhang Z, Wang M. The responses of the gut microbiota to MBL deficiency. Mol Immunol 2020; 122:99-108. [PMID: 32330757 DOI: 10.1016/j.molimm.2020.03.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/09/2020] [Accepted: 03/12/2020] [Indexed: 02/07/2023]
Abstract
Mannose-binding lectin (MBL) deficiency is a common innate immune system deficiency, and is associated with exacerbations and increased colonization of some pathogens. However, the response of the gut microbiota, a pivotal factor in host health, to MBL deficiency is not clear. In this study, MBL-/- and wild-type (WT) mice were generated by backcrossing from MBL-A and MBL-C knockout (KO) mice, and fecal samples were collected at different ages (4th, 8th, 12th, 19th and 27th weeks). The gut microbiota was analyzed by high-throughput sequencing with universal 16S rDNA primers (V3-V5 region). The results showed that structural segregation of the gut microbiota occurred at the 8th, 12th, 19th and 27th weeks of age, although there were no significant differences in alpha diversities between MBL-/- and WT mice at different ages. Impressively, in MBL-/- mice, Akkermansia (from the family Verrucomicrobiaceae) were decreased significantly, Lactobacillus (from the family Lactobacillaceae) abundances, Alistipes and Rikenella (both from the family of Rikenellaceae) were always enriched. Network analysis showed that more interactions existed in the gut microbiota from WT mice (33 nodes and 70 edges) than in the gut microbiota from MBL-/- mice (23 nodes and 40 edges). The 16S rDNA function prediction results indicated that the abundances of predicted genes in the "immune system disease", "metabolic disease" and "nucleotide metabolism" pathways were significantly increased in the MBL-/- mice. In conclusion, this study revealed that the gut microbiota changed in MBL deficient mice, especially at ages older than 4 weeks.
Collapse
Affiliation(s)
- Minna Wu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Fanping Wang
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Immunoregulation and Molecular Diagnostics, Xinxiang 453003, China
| | - Jingwen Yang
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Immunoregulation and Molecular Diagnostics, Xinxiang 453003, China
| | - Puze Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Dong Yan
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Yonghui Yang
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Immunoregulation and Molecular Diagnostics, Xinxiang 453003, China
| | - Wei Zhang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Jie Ren
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Zhenchao Zhang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Mingyong Wang
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Immunoregulation and Molecular Diagnostics, Xinxiang 453003, China.
| |
Collapse
|
35
|
Matsuzaki K, Iwai K, Yoshikawa Y, Shimamura Y, Miyoshi N, Hiramoto S, Asada K, Fukutomi R, Su H, Ohashi N. Wheat Bran Intake Enhances the Secretion of Bacteria-Binding IgA in a Lumen of the Intestinal Tract by Incrementing Short Chain Fatty Acid Production Through Modulation of Gut Microbiota. Nat Prod Commun 2020. [DOI: 10.1177/1934578x20917791] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Wheat bran, a by-product generated in large amounts during wheat processing, consists of 36.5% to 52.4% total dietary fiber. In this study, we investigated the effects of wheat bran intake on the intestinal tract immune system through the modulation of gut microbiota. Balb/c mice were fed with AIN-93G diets containing wheat bran with 2 different particle sizes (average particle size of 53 µm: powdered wheat bran; PWB, and 350 µm: granulated wheat bran; WB) as dietary fibers for 4 weeks. In the wheat bran intake groups, short chain fatty acids (SCFAs: acetic acid, propionic acid, and butyric acid) in the feces were increased after the intake of both particle-size diets, especially in the PWB group, in which the increase occurred immediately. 16S rRNA-based metagenomics of the fecal microbiota revealed that the Shannon Index (α-diversity) and weighted UniFrac distances (β-diversity) in wheat bran intake groups were significantly higher than those in the Control group, and the ratio of the certain family within the order Clostridiales in the fecal microbiota was increased after wheat bran intake, probably some including SCFA-producing bacteria. CXCR5, which is a key surface marker expressed on T follicular helper (Tfh) cells, tended to increase at the expression level in wheat bran intake groups. In addition, the amounts of secretory immunoglobulin A (IgA) and the proportion of IgA-binding bacteria in the feces from wheat bran intake groups were significantly higher than those from the Control group. These findings suggest that wheat bran may enhance Tfh-mediated IgA production in the intestine by SCFA increment through the modulation of gut microbiota and is expected to maintain and improve a healthy intestinal environment.
Collapse
Affiliation(s)
- Konosuke Matsuzaki
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Japan
| | - Katsuki Iwai
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Japan
| | - Yuko Yoshikawa
- School of Veterinary Medicine, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Yuko Shimamura
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Japan
| | - Noriyuki Miyoshi
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Japan
| | | | - Kenichi Asada
- Health Care Research Center, Nisshin Pharma Inc., Tokyo, Japan
| | - Ryuuta Fukutomi
- Health Care Research Center, Nisshin Pharma Inc., Tokyo, Japan
| | - Hongru Su
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Japan
| | - Norio Ohashi
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Japan
| |
Collapse
|
36
|
Abstract
Understanding how health-promoting microbiota are established and their beneficial interactions with the host is of critical biomedical importance. The current high throughput data acquisition technologies allow for integrating components of the gut ecosystem. The richness of data types and large number of measured variables involved underscores the critical importance of the appropriate choice of analytical and computational methods that can be used to model this complex ecosystem. This review outlines currently used approaches to perform analyses of data obtained as a result of interrogation of the gut-microbiota ecosystem and the challenges associated with these methodological and computational efforts. The problem of large dimensionality versus small numbers of samples is explained with discussions of clustering, dimensionality reduction, and statistical testing. Predictive modeling and data integration specific to the gut ecosystem are also discussed.
Collapse
Affiliation(s)
- Ivan Ivanov
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, USA.
| |
Collapse
|
37
|
Ahern PP, Maloy KJ. Understanding immune-microbiota interactions in the intestine. Immunology 2020; 159:4-14. [PMID: 31777071 PMCID: PMC6904641 DOI: 10.1111/imm.13150] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/01/2019] [Accepted: 10/07/2019] [Indexed: 12/13/2022] Open
Abstract
The past two decades have seen an explosion in research that aims to understand how the dynamic interplay with the gut microbiota impacts host health and disease, establishing a role for the gut microbiota in a plethora of pathologies. Understanding how health-promoting microbiota are established and how beneficial host-microbiota interactions are maintained is of immense biomedical importance. Despite the enormous progress that has been made, our knowledge of the specific microbiota members that mediate these effects and the mechanisms underlying these interactions is rudimentary. The dearth of information regarding the nature of advantageous host-microbiota interactions, and the factors that cause these relationships to go awry, has hampered our ability to realize the therapeutic potential of the microbiota. Here we discuss key issues that limit current knowledge and describe a path forwards to improving our understanding of the contributions of the microbiota to host health.
Collapse
Affiliation(s)
- Philip P. Ahern
- Department of Cardiovascular and Metabolic SciencesCleveland Clinic Lerner Research InstituteClevelandOHUSA
| | - Kevin J. Maloy
- Institute of Infection, Immunity and InflammationCollege of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| |
Collapse
|
38
|
Metcalf CJE, Koskella B. Protective microbiomes can limit the evolution of host pathogen defense. Evol Lett 2019; 3:534-543. [PMID: 31636945 PMCID: PMC6791398 DOI: 10.1002/evl3.140] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 08/22/2019] [Accepted: 08/28/2019] [Indexed: 12/17/2022] Open
Abstract
The evolution of host immunity occurs in the context of the microbiome, but little theory exists to predict how resistance against pathogens might be influenced by the need to tolerate and regulate commensal microbiota. We present a general model to explore the optimal investment in host immunity under conditions in which the host can, versus cannot easily distinguish among commensal versus pathogenic bacteria, and when commensal microbiota can, versus cannot protect the host against the impacts of pathogen infection. We find that a loss of immune vigilance associated with innate immunity over evolutionary time can occur due to the challenge of discriminating between pathogenic and other microbe species. Further, we find the greater the protective effect of microbiome species, acting either directly or via competition with a pathogen, or the higher the costs of immunity, the more likely the loss of immune vigilance is. Conversely, this effect can be reversed when pathogens increase host mortality. Generally, the magnitude of costs of immunity required to allow evolution of decreased immune vigilance are predicted to be lowest when microbiome and pathogen species most resemble each other (in terms of host recognition), and when immune effects on the pathogen are weak. Our model framework makes explicit the core trade‐offs likely to shape the evolution of immunity in the context of microbiome/pathogen discrimination. We discuss how this informs interpretation of patterns and process in natural systems, including vulnerability to pathogen emergence.
Collapse
Affiliation(s)
- C Jessica E Metcalf
- Department of Ecology and Evolutionary Princeton University Princeton New Jersey 08540
| | - Britt Koskella
- Department of Integrative Biology University of California Berkeley Berkeley California 94720
| |
Collapse
|
39
|
Lanzoni O, Plotnikov A, Khlopko Y, Munz G, Petroni G, Potekhin A. The core microbiome of sessile ciliate Stentor coeruleus is not shaped by the environment. Sci Rep 2019; 9:11356. [PMID: 31388025 PMCID: PMC6684585 DOI: 10.1038/s41598-019-47701-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 07/22/2019] [Indexed: 12/29/2022] Open
Abstract
Microbiomes of multicellular organisms are one of the hottest topics in microbiology and physiology, while only few studies addressed bacterial communities associated with protists. Protists are widespread in all environments and can be colonized by plethora of different bacteria, including also human pathogens. The aim of this study was to characterize the prokaryotic community associated with the sessile ciliate Stentor coeruleus. 16S rRNA gene metabarcoding was performed on single cells of S. coeruleus and on their environment, water from the sewage stream. Our results showed that the prokaryotic community composition differed significantly between Stentor cells and their environment. The core microbiome common for all ciliate specimens analyzed could be defined, and it was composed mainly by representatives of bacterial genera which include also potential human pathogens and commensals, such as Neisseria, Streptococcus, Capnocytophaga, Porphyromonas. Numerous 16S rRNA gene contigs belonged to endosymbiont “Candidatus Megaira polyxenophila”. Our data suggest that each ciliate cell can be considered as an ecological microniche harboring diverse prokaryotic organisms. Possible benefits for persistence and transmission in nature for bacteria associated with protists are discussed. Our results support the hypothesis that ciliates attract potentially pathogenic bacteria and play the role of natural reservoirs for them.
Collapse
Affiliation(s)
| | - Andrey Plotnikov
- Center of Shared Scientific Equipment, Institute for Cellular and Intracellular Symbiosis, Ural Division of RAS, Orenburg, Russia
| | - Yuri Khlopko
- Center of Shared Scientific Equipment, Institute for Cellular and Intracellular Symbiosis, Ural Division of RAS, Orenburg, Russia
| | - Giulio Munz
- Department of Civil and Environmental Engineering, University of Florence, Florence, Italy
| | | | - Alexey Potekhin
- Faculty of Biology, Saint Petersburg State University, Saint Petersburg, Russia.
| |
Collapse
|
40
|
Ruff WE, Dehner C, Kim WJ, Pagovich O, Aguiar CL, Yu AT, Roth AS, Vieira SM, Kriegel C, Adeniyi O, Mulla MJ, Abrahams VM, Kwok WW, Nussinov R, Erkan D, Goodman AL, Kriegel MA. Pathogenic Autoreactive T and B Cells Cross-React with Mimotopes Expressed by a Common Human Gut Commensal to Trigger Autoimmunity. Cell Host Microbe 2019; 26:100-113.e8. [PMID: 31227334 PMCID: PMC8194364 DOI: 10.1016/j.chom.2019.05.003] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 04/08/2019] [Accepted: 05/08/2019] [Indexed: 12/21/2022]
Abstract
Given the immense antigenic load present in the microbiome, we hypothesized that microbiota mimotopes can be a persistent trigger in human autoimmunity via cross-reactivity. Using antiphospholipid syndrome (APS) as a model, we demonstrate cross-reactivity between non-orthologous mimotopes expressed by a common human gut commensal, Roseburia intestinalis (R. int), and T and B cell autoepitopes in the APS autoantigen β2-glycoprotein I (β2GPI). Autoantigen-reactive CD4+ memory T cell clones and an APS-derived, pathogenic monoclonal antibody cross-reacted with R. int mimotopes. Core-sequence-dependent anti-R. int mimotope IgG titers were significantly elevated in APS patients and correlated with anti-β2GPI IgG autoantibodies. R. int immunization of mice induced β2GPI-specific lymphocytes and autoantibodies. Oral gavage of susceptible mice with R. int induced anti-human β2GPI autoantibodies and autoimmune pathologies. Together, these data support a role for non-orthologous commensal-host cross-reactivity in the development and persistence of autoimmunity in APS, which may apply more broadly to human autoimmune disease.
Collapse
Affiliation(s)
- William E Ruff
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Carina Dehner
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Woo J Kim
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Odelya Pagovich
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Cassyanne L Aguiar
- Barbara Volcker Center for Women and Rheumatic Diseases, Hospital for Special Surgery, Weill Cornell Medicine, New York, NY 10021, USA
| | - Andrew T Yu
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Alexander S Roth
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | | | - Christina Kriegel
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Olamide Adeniyi
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Melissa J Mulla
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA
| | - Vikki M Abrahams
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA
| | - William W Kwok
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - Ruth Nussinov
- Computational Structural Biology Section, Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; Sackler Institute of Molecular Medicine, Department of Human Genetics and Molecular Medicine, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Doruk Erkan
- Barbara Volcker Center for Women and Rheumatic Diseases, Hospital for Special Surgery, Weill Cornell Medicine, New York, NY 10021, USA
| | - Andrew L Goodman
- Microbial Sciences Institute, Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT 06536, USA
| | - Martin A Kriegel
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA; Department of Medicine, Section of Rheumatology, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
41
|
The Gut Microbiome in Inflammatory Bowel Disease: Lessons Learned From Other Immune-Mediated Inflammatory Diseases. Am J Gastroenterol 2019; 114:1051-1070. [PMID: 31232832 DOI: 10.14309/ajg.0000000000000305] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There is a growing appreciation for the role of the gut microbiome in human health and disease. Aided by advances in sequencing technologies and analytical methods, recent research has shown the healthy gut microbiome to possess considerable diversity and functional capacity. Dysbiosis of the gut microbiota is believed to be involved in the pathogenesis of not only diseases that primarily affect the gastrointestinal tract but also other less obvious diseases, including neurologic, rheumatologic, metabolic, hepatic, and other illnesses. Chronic immune-mediated inflammatory diseases (IMIDs) represent a group of diseases that share many underlying etiological factors including genetics, aberrant immunological responses, and environmental factors. Gut dysbiosis has been reported to be common to IMIDs as a whole, and much effort is currently being directed toward elucidating microbiome-mediated disease mechanisms and their implications for causality. In this review, we discuss gut microbiome studies in several IMIDs and show how these studies can inform our understanding of the role of the gut microbiome in inflammatory bowel disease.
Collapse
|
42
|
Stebegg M, Silva-Cayetano A, Innocentin S, Jenkins TP, Cantacessi C, Gilbert C, Linterman MA. Heterochronic faecal transplantation boosts gut germinal centres in aged mice. Nat Commun 2019; 10:2443. [PMID: 31164642 PMCID: PMC6547660 DOI: 10.1038/s41467-019-10430-7] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 05/10/2019] [Indexed: 12/21/2022] Open
Abstract
Ageing is a complex multifactorial process associated with a plethora of disorders, which contribute significantly to morbidity worldwide. One of the organs significantly affected by age is the gut. Age-dependent changes of the gut-associated microbiome have been linked to increased frailty and systemic inflammation. This change in microbial composition with age occurs in parallel with a decline in function of the gut immune system; however, it is not clear whether there is a causal link between the two. Here we report that the defective germinal centre reaction in Peyer's patches of aged mice can be rescued by faecal transfers from younger adults into aged mice and by immunisations with cholera toxin, without affecting germinal centre reactions in peripheral lymph nodes. This demonstrates that the poor germinal centre reaction in aged animals is not irreversible, and that it is possible to improve this response in older individuals by providing appropriate stimuli.
Collapse
Affiliation(s)
- Marisa Stebegg
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Alyssa Silva-Cayetano
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Silvia Innocentin
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Timothy P Jenkins
- Department of Veterinary Medicine, Madingley Road, Cambridge, CB3 0ES, UK
| | - Cinzia Cantacessi
- Department of Veterinary Medicine, Madingley Road, Cambridge, CB3 0ES, UK
| | - Colin Gilbert
- Biological Services Unit, Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Michelle A Linterman
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK.
| |
Collapse
|
43
|
Cross talk between neutrophils and the microbiota. Blood 2019; 133:2168-2177. [PMID: 30898860 DOI: 10.1182/blood-2018-11-844555] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 12/24/2018] [Indexed: 12/18/2022] Open
Abstract
The microbiota has emerged as an important regulator of the host immunity by the induction, functional modulation, or suppression of local and systemic immune responses. In return, the host immune system restricts translocation and fine tunes the composition and distribution of the microbiota to maintain a beneficial symbiosis. This paradigm applies to neutrophils, a critical component of the innate immunity, allowing their production and function to be influenced by microbial components and metabolites derived from the microbiota, and engaging them in the process of microbiota containment and regulation. The cross talk between neutrophils and the microbiota adjusts the magnitude of neutrophil-mediated inflammation on challenge while preventing neutrophil responses against commensals under steady state. Here, we review the major molecular and cellular mediators of the interactions between neutrophils and the microbiota and discuss their interplay and contribution in chronic inflammatory diseases and cancer.
Collapse
|
44
|
Gut microbiota, a new frontier to understand traditional Chinese medicines. Pharmacol Res 2019; 142:176-191. [PMID: 30818043 DOI: 10.1016/j.phrs.2019.02.024] [Citation(s) in RCA: 253] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/18/2019] [Accepted: 02/23/2019] [Indexed: 02/06/2023]
Abstract
As an important component of complementary and alternative medicines, traditional Chinese medicines (TCM) are gaining more and more attentions around the world because of the powerful therapeutic effects and less side effects. However, there are still some doubts about TCM because of the questionable TCM theories and unclear biological active compounds. In recent years, gut microbiota has emerged as an important frontier to understand the development and progress of diseases. Together with this trend, an increasing number of studies have indicated that drug molecules can interact with gut microbiota after oral administration. In this context, more and more studies pertaining to TCM have paid attention to gut microbiota and have yield rich information for understanding TCM. After oral administration, TCM can interact with gut microbiota: (1) TCM can modulate the composition of gut microbiota; (2) TCM can modulate the metabolism of gut microbiota; (3) gut microbiota can transform TCM compounds. During the interactions, two types of metabolites can be produced: gut microbiota metabolites (of food and host origin) and gut microbiota transformed TCM compounds. In this review, we summarized the interactions between TCM and gut microbiota, and the pharmacological effects and features of metabolites produced during interactions between TCM and gut microbiota. Then, focusing on gut microbiota and metabolites, we summarized the aspects in which gut microbiota has facilitated our understanding of TCM. At the end of this review, the outlooks for further research of TCM and gut microbiota were also discussed.
Collapse
|
45
|
Affiliation(s)
- Catherine S. Birch
- Institute for Agri‐Food Research & Innovation, School of Natural & Environmental Sciences Newcastle University Newcastle NE1 7RU UK
| | - Graham A. Bonwick
- Institute for Agri‐Food Research & Innovation, School of Natural & Environmental Sciences Newcastle University Newcastle NE1 7RU UK
- Fera Science Limited Sand Hutton, York YO41 1LZ UK
| |
Collapse
|
46
|
Feng W, Ao H, Peng C. Gut Microbiota, Short-Chain Fatty Acids, and Herbal Medicines. Front Pharmacol 2018; 9:1354. [PMID: 30532706 PMCID: PMC6265305 DOI: 10.3389/fphar.2018.01354] [Citation(s) in RCA: 239] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 11/05/2018] [Indexed: 12/12/2022] Open
Abstract
As an important source for traditional medical systems such as Ayurvedic medicine and traditional Chinese medicine, herbal medicines have received widespread attentions from all over the world, especially in developing countries. Over the past decade, studies on gut microbiota have generated rich information for understanding how gut microbiota shape the functioning of our body system. In view of the importance of gut microbiota, the researchers engaged in studying herbal medicines have paid more and more attention to gut microbiota and gut microbiota metabolites. Among a variety of gut microbiota metabolites, short-chain fatty acids (SCFAs) have received most attention because of their important role in maintaining the hemostasis of hosts and recovery of diseases. Herbal medicines, as an important resource provider for production of SCFAs, have been demonstrated to be able to modulate gut microbiota composition and regulate SCFAs production. In this mini-review, we summarize current knowledge about SCFAs origination, the role of SCFAs in health and disease, the influence of herbal medicine on SCFAs production and the corresponding mechanisms. At the end of this review, the strategies and suggestions for further research of SCFAs and herbal medicines are also discussed.
Collapse
Affiliation(s)
- Wuwen Feng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hui Ao
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
47
|
Myhill LJ, Stolzenbach S, Hansen TVA, Skovgaard K, Stensvold CR, Andersen LO, Nejsum P, Mejer H, Thamsborg SM, Williams AR. Mucosal Barrier and Th2 Immune Responses Are Enhanced by Dietary Inulin in Pigs Infected With Trichuris suis. Front Immunol 2018; 9:2557. [PMID: 30473696 PMCID: PMC6237860 DOI: 10.3389/fimmu.2018.02557] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 10/17/2018] [Indexed: 12/12/2022] Open
Abstract
Diet composition may play a crucial role in shaping host immune responses and commensal gut microbiota populations. Bioactive dietary components, such as inulin, have been extensively studied for their bioactive properties, particularly in modulating gut immune function and reducing inflammation. It has been shown that colonization with gastrointestinal parasitic worms (helminths) may alleviate chronic inflammation through promotion of T-helper cell type (Th) 2 and T-regulatory immune responses and alterations in the gut microbiome. In this study, we investigated if dietary inulin could modulate mucosal immune function in pigs during colonization with the porcine whipworm Trichuris suis. T. suis infection induced a typical Th2-biased immune response characterized by transcriptional changes in Th2- and barrier function-related genes, accompanied by intestinal remodeling through increased epithelial goblet and tuft cell proliferation. We observed that inulin also up-regulated Th2-related immune genes (IL13, IL5), and suppressed Th1-related pro-inflammatory genes (IFNG, IL1A, IL8) in the colon. Notably, inulin augmented the T. suis-induced responses with increased transcription of key Th2 and mucosal barrier genes (e.g., IL13, TFF3), and synergistically suppressed pro-inflammatory genes, such as IFNG and CXCL9. 16S rRNA sequencing of proximal colon digesta samples revealed that inulin supplementation reduced the abundance of bacterial phyla linked to inflammation, such as Proteobacteria and Firmicutes, and simultaneously increased Actinobacteria and Bacteroidetes. Interestingly, pigs treated with both inulin and T. suis displayed the highest Bacteroidetes: Firmicutes ratio and the lowest gut pH, suggesting an interaction of diet and helminth infection that stimulates the growth of beneficial bacterial species. Overall, our data demonstrate that T. suis infection and inulin co-operatively enhance anti-inflammatory immune responses, which is potentially mediated by changes in microbiota composition. Our results highlight the intricate interactions between diet, immune function and microbiota composition in a porcine helminth infection model. This porcine model should facilitate further investigations into the use of bioactive diets as immunomodulatory mediators against inflammatory conditions, and how diet and parasites may influence gut health.
Collapse
Affiliation(s)
- Laura J Myhill
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sophie Stolzenbach
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tina V A Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kerstin Skovgaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - C Rune Stensvold
- Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Lee O'Brien Andersen
- Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Peter Nejsum
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Helena Mejer
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stig M Thamsborg
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andrew R Williams
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|