1
|
Chen J, Pan Q, Lu L, Huang X, Wang S, Liu X, Lun J, Xu X, Su H, Guo F, Yang L, You L, Xiao H, Luo W, Liu HF, Pan Q. Atg5 deficiency in basophils improves metabolism in lupus mice by regulating gut microbiota dysbiosis. Cell Commun Signal 2025; 23:40. [PMID: 39844180 PMCID: PMC11756211 DOI: 10.1186/s12964-025-02041-1] [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: 11/10/2024] [Accepted: 01/12/2025] [Indexed: 01/24/2025] Open
Abstract
Autophagic activation in immune cells, gut microbiota dysbiosis, and metabolic abnormalities have been reported separately as characteristics of systemic lupus erythematosus (SLE). Elucidating the crosstalk among the immune system, commensal microbiota, and metabolites is crucial to understanding the pathogenesis of autoimmune diseases. Emerging evidence shows that basophil activation plays a critical role in the pathogenesis of SLE; however, the underlying mechanisms remain largely unknown. Here, we investigated the effects of autophagic inhibition on the pathogenesis of basophils in SLE using Autophagy-related gene 5 (Atg5) knockout (Atg5-/-) as an autophagic inhibitor. Specifically, we knocked out basophilic Atg5 in vivo to investigate its impact on lupus metabolism. Furthermore, Atg5-/- basophils were transferred to basophil-depleted MRL/MpJ-Faslpr (MRL/lpr) mice to study their effect on disease metabolism. Metagenomic and targeted metabolomic sequencing results indicated considerable reduction in the levels of plasma autoantibodies and inflammatory cytokines in the Atg5-/- basophil transfer group compared with that in the control group. Transplanting Atg5-/- basophils improved the gut microbiota balance in MRL/lpr mice, increasing the abundance of beneficial bacteria, such as Ligilactobacillus murinus and Faecalitalea rodentium, and reducing that of potentially pathogenic bacteria such as Phocaeicola salanitronis. The transplantation of Atg5-deficient basophils improved lupus symptoms by modulating lipid and amino acid metabolism. This improvement was linked to changes in the gut microbiota, particularly an increase in Ligilactobacillus murinus and Faecalitalea rodentium populations. These microbial shifts are believed to promote the production of beneficial metabolites, such as γ-linolenic acid and oleoyl-1-palmitoyl-sn-glycero-3-phosphocholine, while reducing the levels of harmful metabolites such as arginine. These alterations in the metabolic profile contribute to the alleviation of lupus symptoms. Collectively, these findings reveal a novel role of basophil autophagy in SLE, highlighting its potential as a therapeutic target.
Collapse
Affiliation(s)
- Jiaxuan Chen
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Quanren Pan
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lu Lu
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiaorong Huang
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shuting Wang
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiaoxian Liu
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jiaqi Lun
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiaowei Xu
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Hongyong Su
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Fengbiao Guo
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lawei Yang
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Liuyong You
- Department of Clinical Laboratory, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Haiyan Xiao
- Department of Cellular Biology and Anatomy, James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Wenying Luo
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Hua-Feng Liu
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
| | - Qingjun Pan
- Department of Nephrology, National Clinical Key Specialty Construction Program, Institute of Nephrology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
- Department of Clinical Laboratory, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
2
|
Liu Y, Yang X. A review on the novel biomarkers of systemic lupus erythematosus discovered via metabolomic profiling. Front Immunol 2024; 15:1443440. [PMID: 39569194 PMCID: PMC11576423 DOI: 10.3389/fimmu.2024.1443440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 10/21/2024] [Indexed: 11/22/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a multifaceted autoimmune disease affecting various body organs and systems. The diagnosis of SLE and its complications is based on evident clinical symptoms, serological marker levels, and pathological findings. Some serological markers have a low sensitivity and specificity, and biopsy procedures are invasive in nature. Hence, metabolomics has emerged as a valuable tool for SLE screening and categorization. Its application has contributed significantly to identifying SLE pathogenesis, improving clinical diagnosis, and developing treatment approaches. This review provides an overview of the utilization of metabolomics in the study of SLE, focusing on advancements in understanding the disease's pathogenesis, aiding in diagnosis, and monitoring treatment efficacy.
Collapse
Affiliation(s)
- Yinghong Liu
- Department of Rheumatology, Chongqing University Central Hospital, Chongqing, China
- Department of Rheumatology, Chongqing Emergency Medical Center, Chongqing, China
| | - Xiaojuan Yang
- Department of Rheumatology, Chongqing University Central Hospital, Chongqing, China
- Department of Rheumatology, Chongqing Emergency Medical Center, Chongqing, China
| |
Collapse
|
3
|
Su X, Wang X, Zhang X, Sun Y, Jia Y. β-Indole-3-acetic acid attenuated collagen-induced arthritis through reducing the ubiquitination of Foxp3 via the AhR-TAZ-Tip60 pathway. Immunol Res 2024; 72:741-753. [PMID: 38630408 DOI: 10.1007/s12026-024-09480-x] [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: 02/09/2024] [Accepted: 04/09/2024] [Indexed: 08/28/2024]
Abstract
Massive evidence shows that intestinal tryptophan metabolites affected by intestinal flora can modulate the progression of rheumatoid arthritis (RA). However, the effects and mechanisms of intestinal tryptophan metabolites on RA are not yet detailed. Herein, we investigated the protective effects of intestinal tryptophan metabolites on RA and its detailed mechanisms. In this study, the collagen-induced arthritis (CIA) rat model was established. Based on metabolomics analysis, the contents of β-indole-3-acetic acid (IAA), indolylpropionic acid, and indole-3-β-acrylic acid in the sera of CIA rats were significantly less compared with those of the normal rats. Under the condition of Treg or Th17 cell differentiation, IAA significantly promoted the differentiation and activation of Treg cells instead of Th17 cells. Intestinal tryptophan metabolites are well-known endogenic ligands of aryl hydrocarbon receptor (AhR). Not surprisingly, IAA increased the level of Foxp3 through activating the AhR pathway. Interestingly, IAA had little impact on the level of Foxp3 mRNA, but reducing the ubiquitination and degradation of Foxp3. Mechanically, IAA reduced the expression of the transcriptional coactivator TAZ, which was almost completely reversed by either AhR antagonist CH223191 or siRNA. In vitro, IAA decreased the combination of TAZ and the histone acetyltransferase Tip60, while it increased the combination of Tip60 and Foxp3. In CIA rats, oral administration of IAA increased the number of Treg cells and relieved the inflammation. A combined use with CH223191 almost abolished the effect of IAA. Taken together, IAA attenuated CIA by promoting the differentiation of Treg cells through reducing the ubiquitination of Foxp3 via the AhR-TAZ-Tip60 pathway.
Collapse
Affiliation(s)
- Xiaoran Su
- Department of Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Xinliu Wang
- Department of Integrative Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Xin Zhang
- Department of Integrative Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Yajie Sun
- Department of Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Yugai Jia
- Department of Pharmacology, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China.
- Hebei International Cooperation Center for Ion Channel Function and Innovative Traditional Chinese Medicine, Shijiazhuang, 050091, China.
| |
Collapse
|
4
|
Zhao M, Wen X, Liu R, Xu K. Microbial dysbiosis in systemic lupus erythematosus: a scientometric study. Front Microbiol 2024; 15:1319654. [PMID: 38863759 PMCID: PMC11166128 DOI: 10.3389/fmicb.2024.1319654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 05/01/2024] [Indexed: 06/13/2024] Open
Abstract
Introduction Systemic lupus erythematosus (SLE) is a chronic autoimmune disease. Mounting evidence suggests microbiota dysbiosis augment autoimmune response. This study aims to provide a systematic overview of this research field in SLE through a bibliometric analysis. Methods We conducted a comprehensive search and retrieval of literature related to microbial researches in SLE from the Web of Science Core Collection (WOSCC) database. The retrieved articles were subjected to bibliometric analysis using VOSviewer and Bibliometricx to explore annual publication output, collaborative patterns, research hotspots, current research status, and emerging trends. Results In this study, we conducted a comprehensive analysis of 218 research articles and 118 review articles. The quantity of publications rises annually, notably surging in 2015 and 2018. The United States and China emerged as the leading contributors in microbial research of SLE. Mashhad University of Medical Sciences had the highest publication outputs among the institutions. Frontiers in Immunology published the most papers. Luo XM and Margolles A were the most prolific and highly cited contributors among individual authors. Microbial research in SLE primarily focused on changes in microbial composition, particularly gut microbiota, as well as the mechanisms and practical applications in SLE. Recent trends emphasize "metabolites," "metabolomics," "fatty acids," "T cells," "lactobacillus," and "dietary supplementation," indicating a growing emphasis on microbial metabolism and interventions in SLE. Conclusion This study provides a thorough analysis of the research landscape concerning microbiota in SLE. The microbial research in SLE mainly focused on three aspects: microbial dysbiosis, mechanism studies and translational studies (microbiota-based therapeutics). It identifies current research trends and focal points, offering valuable guidance for scholars in the field.
Collapse
Affiliation(s)
- Miaomiao Zhao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Xiaoting Wen
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Ruiling Liu
- Department of Microbiology and Immunology, Basic Medical College, Shanxi Medical University, Jinzhong, China
| | - Ke Xu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
5
|
Han D, Wang W, Gong J, Ma Y, Li Y. Microbiota metabolites in bone: Shaping health and Confronting disease. Heliyon 2024; 10:e28435. [PMID: 38560225 PMCID: PMC10979239 DOI: 10.1016/j.heliyon.2024.e28435] [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: 12/03/2023] [Revised: 02/16/2024] [Accepted: 03/19/2024] [Indexed: 04/04/2024] Open
Abstract
The intricate interplay between the gut microbiota and bone health has become increasingly recognized as a fundamental determinant of skeletal well-being. Microbiota-derived metabolites play a crucial role in dynamic interaction, specifically in bone homeostasis. In this sense, short-chain fatty acids (SCFAs), including acetate, propionate, and butyrate, indirectly promote bone formation by regulating insulin-like growth factor-1 (IGF-1). Trimethylamine N-oxide (TMAO) has been found to increase the expression of osteoblast genes, such as Runt-related transcription factor 2 (RUNX2) and bone morphogenetic protein-2 (BMP2), thus enhancing osteogenic differentiation and bone quality through BMP/SMADs and Wnt signaling pathways. Remarkably, in the context of bone infections, the role of microbiota metabolites in immune modulation and host defense mechanisms potentially affects susceptibility to infections such as osteomyelitis. Furthermore, ongoing research elucidates the precise mechanisms through which microbiota-derived metabolites influence bone cells, such as osteoblasts and osteoclasts. Understanding the multifaceted influence of microbiota metabolites on bone, from regulating homeostasis to modulating susceptibility to infections, has the potential to revolutionize our approach to bone health and disease management. This review offers a comprehensive exploration of this evolving field, providing a holistic perspective on the impact of microbiota metabolites on bone health and diseases.
Collapse
Affiliation(s)
- Dong Han
- Department of Trauma Orthopedics, Yantaishan Hospital, Yantai 264000, China
| | - Weijiao Wang
- Department of Otolaryngology, Yantaishan Hospital, Yantai 264000, China
| | - Jinpeng Gong
- Department of Trauma Orthopedics, Yantaishan Hospital, Yantai 264000, China
| | - Yupeng Ma
- Department of Trauma Orthopedics, Yantaishan Hospital, Yantai 264000, China
| | - Yu Li
- Department of Trauma Orthopedics, Yantaishan Hospital, Yantai 264000, China
| |
Collapse
|
6
|
Bhattacharjee P, Karim KA, Khan Z. Harnessing the Microbiome: A Comprehensive Review on Advancing Therapeutic Strategies for Rheumatic Diseases. Cureus 2023; 15:e50964. [PMID: 38249228 PMCID: PMC10800157 DOI: 10.7759/cureus.50964] [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] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
Rheumatic diseases are a group of disorders that affect the joints, muscles, and bones. These diseases, such as rheumatoid arthritis, lupus, and psoriatic arthritis, can cause pain, stiffness, and swelling, leading to reduced mobility and disability. Recent studies have identified the microbiome, the diverse community of microorganisms that live in and on the human body, as a potential factor in the development and progression of rheumatic diseases. Harnessing the microbiome offers a promising new avenue for developing therapeutic strategies for these debilitating conditions. There is growing interest in the role of oral and gut microbiomes in the management of rheumatoid arthritis and other autoimmune disease. Microbial metabolites have immunomodulatory properties that could be exploited for rheumatic disorders. A wide range of microorganisms are present in the oral cavity and are found to be vulnerable to the effects of the environment. The physiology and ecology of the microbiota become intimately connected with those of the host, and they critically influence the promotion of health or progression toward disease. This article aims to provide a comprehensive overview of the current state of knowledge on oral and gut microbiome and its potential future role in the management of rheumatic diseases. This article will also discuss newer treatment strategies such as bioinformatic analyses and fecal transplantation.
Collapse
Affiliation(s)
- Priyadarshini Bhattacharjee
- Acute Medicine, Cambridge University Hospital NHS Foundation Trust, Cambridge, GBR
- School of Clinical Medicine, University of Cambridge, Cambridge, GBR
| | - Karim Arif Karim
- Medicine and Surgery, Kamuzu University of Health Sciences, Blantyre, MWI
| | - Zahid Khan
- Acute Medicine, Mid and South Essex NHS Foundation Trust, Southend-on-Sea, GBR
- Cardiology, Bart's Heart Centre, London, GBR
- Cardiology and General Medicine, Barking, Havering and Redbridge University Hospitals NHS Trust, London, GBR
- Cardiology, Royal Free Hospital, London, GBR
| |
Collapse
|
7
|
Luo Y, Tong Y, Wu L, Niu H, Li Y, Su LC, Wu Y, Bozec A, Zaiss MM, Qing P, Zhao H, Tan C, Zhang Q, Zhao Y, Tang H, Liu Y. Alteration of Gut Microbiota in Individuals at High-Risk for Rheumatoid Arthritis Associated With Disturbed Metabolome and the Initiation of Arthritis Through the Triggering of Mucosal Immunity Imbalance. Arthritis Rheumatol 2023; 75:1736-1748. [PMID: 37219936 DOI: 10.1002/art.42616] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 04/19/2023] [Accepted: 05/08/2023] [Indexed: 05/24/2023]
Abstract
OBJECTIVE In this study, we aimed to decipher the gut microbiome (GM) and serum metabolic characteristic of individuals at high risk for rheumatoid arthritis (RA) and to investigate the causative effect of GM on the mucosal immune system and its involvement in the pathogenesis of arthritis. METHODS Fecal samples were collected from 38 healthy individuals and 53 high-risk RA individuals with anti-citrullinated protein antibody (ACPA) positivity (Pre-RA), 12 of 53 Pre-RA individuals developed RA within 5 years of follow-up. The differences in intestinal microbial composition between the healthy controls and Pre-RA individuals or among Pre-RA subgroups were identified by 16S ribosomal RNA sequencing. The serum metabolite profile and its correlation with GM were also explored. Moreover, antibiotic-pretreated mice that received GM from the healthy control or Pre-RA groups were then evaluated for intestinal permeability, inflammatory cytokines, and immune cell populations. Collagen-induced arthritis (CIA) was also applied to test the effect of fecal microbiota transplantation (FMT) from Pre-RA individuals on arthritis severity in mice. RESULTS Stool microbial diversity was lower in Pre-RA individuals than in healthy controls. The bacterial community structure and function significantly differed between healthy controls and Pre-RA individuals. Although there were differences to some extent in the bacterial abundance among the Pre-RA subgroups, no robust functional differences were observed. The metabolites in the serum of the Pre-RA group were dramatically different from those in the healthy controls group, with KEGG pathway enrichment of amino acid and lipid metabolism. Moreover, intestinal bacteria from the Pre-RA group increased intestinal permeability in FMT mice and zonula occludens-1 expression in the small intestine and Caco-2 cells. Moreover, Th17 cells in the mesenteric lymph nodes and Peyer's patches were also increased in mice receiving Pre-RA feces compared to healthy controls. The changes in intestinal permeability and Th17-cell activation prior to arthritis induction enhanced CIA severity in PreRA-FMT mice compared with HC-FMT mice. CONCLUSION Gut microbial dysbiosis and metabolome alterations already occur in individuals at high risk for RA. FMT from preclinical individuals triggers intestinal barrier dysfunction and changes mucosal immunity, further contributing to the development of arthritis.
Collapse
Affiliation(s)
- Yubin Luo
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yanli Tong
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Liang Wu
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Haitao Niu
- School of Medicine, Jinan University, Guangzhou, China
| | - Yanhong Li
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Lin Chong Su
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic diseases, Enshi, China
| | - Yuxi Wu
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Aline Bozec
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3-Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Mario M Zaiss
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3-Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Pingying Qing
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Hua Zhao
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Chunyu Tan
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Qiuping Zhang
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Zhao
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Huairong Tang
- Health Management Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Liu
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
8
|
Uceda S, Echeverry-Alzate V, Reiriz-Rojas M, Martínez-Miguel E, Pérez-Curiel A, Gómez-Senent S, Beltrán-Velasco AI. Gut Microbial Metabolome and Dysbiosis in Neurodegenerative Diseases: Psychobiotics and Fecal Microbiota Transplantation as a Therapeutic Approach-A Comprehensive Narrative Review. Int J Mol Sci 2023; 24:13294. [PMID: 37686104 PMCID: PMC10487945 DOI: 10.3390/ijms241713294] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
The comprehensive narrative review conducted in this study delves into the mechanisms of communication and action at the molecular level in the human organism. The review addresses the complex mechanism involved in the microbiota-gut-brain axis as well as the implications of alterations in the microbial composition of patients with neurodegenerative diseases. The pathophysiology of neurodegenerative diseases with neuronal loss or death is analyzed, as well as the mechanisms of action of the main metabolites involved in the bidirectional communication through the microbiota-gut-brain axis. In addition, interventions targeting gut microbiota restructuring through fecal microbiota transplantation and the use of psychobiotics-pre- and pro-biotics-are evaluated as an opportunity to reduce the symptomatology associated with neurodegeneration in these pathologies. This review provides valuable information and facilitates a better understanding of the neurobiological mechanisms to be addressed in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Sara Uceda
- BRABE Group, Psychology Department, School of Life and Nature Sciences, Nebrija University, 28240 Madrid, Spain
| | - Víctor Echeverry-Alzate
- BRABE Group, Psychology Department, School of Life and Nature Sciences, Nebrija University, 28240 Madrid, Spain
| | - Manuel Reiriz-Rojas
- BRABE Group, Psychology Department, School of Life and Nature Sciences, Nebrija University, 28240 Madrid, Spain
| | - Esther Martínez-Miguel
- Health Department, School of Life and Nature Sciences, Nebrija University, 28240 Madrid, Spain
| | - Ana Pérez-Curiel
- Health Department, School of Life and Nature Sciences, Nebrija University, 28240 Madrid, Spain
| | - Silvia Gómez-Senent
- Health Department, School of Life and Nature Sciences, Nebrija University, 28240 Madrid, Spain
| | | |
Collapse
|
9
|
Tsetseri MN, Silman AJ, Keene DJ, Dakin SG. The role of the microbiome in rheumatoid arthritis: a review. Rheumatol Adv Pract 2023; 7:rkad034. [PMID: 38606003 PMCID: PMC11007908 DOI: 10.1093/rap/rkad034] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 03/26/2023] [Indexed: 04/13/2024] Open
Abstract
The close bidirectional relationship between the microbiome and the immune system is well supported, and a role of gut dysbiosis has been implied in many systemic autoimmune diseases. This review aims to provide a critical summary and appraisal of 6 murine studies and 16 clinical studies. The findings of the literature review suggest that gut dysbiosis precedes arthritis and that local intestinal inflammation leads to systemic inflammation in genetically predisposed individuals. However, the exact mechanism by which microorganisms provoke immune responses at distal sites remains to be elucidated. Although a characteristic RA microbiome was not identified, there were some common findings among studies: overabundance of Prevotella copri in early RA patients, and proliferation of the genus Collinsela and some Lactobacillus species. Three mechanisms by which microbiota might contribute to RA pathogenesis were proposed: inflammatory responses (P. copri and Lactobacillus), molecular mimicry (P. copri) and loss of intestinal barrier integrity (Collinsella). Larger longitudinal studies are required in order to shed light on the mechanisms involved and unravel the therapeutic potential of the microbiome, and clinical trials are needed to evaluate the safety and efficacy of the implied therapeutic interventions.
Collapse
Affiliation(s)
- Maria-Nefeli Tsetseri
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Alan J Silman
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - David J Keene
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
- Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Stephanie G Dakin
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| |
Collapse
|
10
|
Fu TC, Kung YY, Lin JR, Chang CM. Editorial: Omics for the objective diagnosis and management of immune-mediated rheumatic diseases. Front Med (Lausanne) 2023; 10:1127430. [PMID: 36714387 PMCID: PMC9880463 DOI: 10.3389/fmed.2023.1127430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 01/02/2023] [Indexed: 01/15/2023] Open
Affiliation(s)
- Tieh-Cheng Fu
- Department of Physical Medicine and Rehabilitation, Chang Gung Memorial Hospital, Keelung, Taiwan,Division of Cardiology, Department of Internal Medicine, Heart Failure Research Center, Chang Gung Memorial Hospital, Keelung, Taiwan,School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yen-Ying Kung
- Center for Traditional Medicine, Taipei Veterans General Hospital, Taipei, Taiwan,Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan,Institute of Traditional Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jr-Rung Lin
- Clinical Informatics and Medical Statistics Research Center, Graduate Institute of Clinical Medical, Chang Gung University, Taoyuan, Taiwan
| | - Ching-Mao Chang
- Center for Traditional Medicine, Taipei Veterans General Hospital, Taipei, Taiwan,Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan,Institute of Traditional Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan,*Correspondence: Ching-Mao Chang ✉
| |
Collapse
|
11
|
Harder JW, Ma J, Alard P, Sokoloski KJ, Mathiowitz E, Furtado S, Egilmez NK, Kosiewicz MM. Male microbiota-associated metabolite restores macrophage efferocytosis in female lupus-prone mice via activation of PPARγ/LXR signaling pathways. J Leukoc Biol 2023; 113:41-57. [PMID: 36822162 DOI: 10.1093/jleuko/qiac002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Indexed: 01/11/2023] Open
Abstract
Systemic lupus erythematosus development is influenced by both sex and the gut microbiota. Metabolite production is a major mechanism by which the gut microbiota influences the immune system, and we have previously found differences in the fecal metabolomic profiles of lupus-prone female and lupus-resistant male BWF1 mice. Here we determine how sex and microbiota metabolite production may interact to affect lupus. Transcriptomic analysis of female and male splenocytes showed genes that promote phagocytosis were upregulated in BWF1 male mice. Because patients with systemic lupus erythematosus exhibit defects in macrophage-mediated phagocytosis of apoptotic cells (efferocytosis), we compared splenic macrophage efferocytosis in vitro between female and male BWF1 mice. Macrophage efferocytosis was deficient in female compared to male BWF1 mice but could be restored by feeding male microbiota. Further transcriptomic analysis of the genes upregulated in male BWF1 mice revealed enrichment of genes stimulated by PPARγ and LXR signaling. Our previous fecal metabolomics analyses identified metabolites in male BWF1 mice that can activate PPARγ and LXR signaling and identified one in particular, phytanic acid, that is a very potent agonist. We show here that treatment of female BWF1 splenic macrophages with phytanic acid restores efferocytic activity via activation of the PPARγ and LXR signaling pathways. Furthermore, we found phytanic acid may restore female BWF1 macrophage efferocytosis through upregulation of the proefferocytic gene CD36. Taken together, our data indicate that metabolites produced by BWF1 male microbiota can enhance macrophage efferocytosis and, through this mechanism, could potentially influence lupus progression.
Collapse
Affiliation(s)
- James W Harder
- Department of Microbiology and Immunology, University of Louisville, 505 South Hancock St, Rm 609, Louisville, KY 40202, USA
| | - Jing Ma
- Department of Microbiology and Immunology, University of Louisville, 505 South Hancock St, Rm 609, Louisville, KY 40202, USA
| | - Pascale Alard
- Department of Microbiology and Immunology, University of Louisville, 505 South Hancock St, Rm 609, Louisville, KY 40202, USA
| | - Kevin J Sokoloski
- Department of Microbiology and Immunology, University of Louisville, 505 South Hancock St, Rm 609, Louisville, KY 40202, USA
| | - Edith Mathiowitz
- Department of Medical Science and Engineering, Brown University, 222 Richmond Street, Providence, RI 02903, USA
| | - Stacia Furtado
- Department of Medical Science and Engineering, Brown University, 222 Richmond Street, Providence, RI 02903, USA
| | - Nejat K Egilmez
- Department of Microbiology and Immunology, University of Louisville, 505 South Hancock St, Rm 609, Louisville, KY 40202, USA
| | - Michele M Kosiewicz
- Department of Microbiology and Immunology, University of Louisville, 505 South Hancock St, Rm 609, Louisville, KY 40202, USA
| |
Collapse
|
12
|
Larabi AB, Masson HLP, Bäumler AJ. Bile acids as modulators of gut microbiota composition and function. Gut Microbes 2023; 15:2172671. [PMID: 36740850 PMCID: PMC9904317 DOI: 10.1080/19490976.2023.2172671] [Citation(s) in RCA: 108] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 01/16/2023] [Indexed: 02/07/2023] Open
Abstract
Changes in the composition of gut-associated microbial communities are associated with many human illnesses, but the factors driving dysbiosis remain incompletely understood. One factor governing the microbiota composition in the gut is bile. Bile acids shape the microbiota composition through their antimicrobial activity and by activating host signaling pathways that maintain gut homeostasis. Although bile acids are host-derived, their functions are integrally linked to bacterial metabolism, which shapes the composition of the intestinal bile acid pool. Conditions that change the size or composition of the bile acid pool can trigger alterations in the microbiota composition that exacerbate inflammation or favor infection with opportunistic pathogens. Therefore, manipulating the composition or size of the bile acid pool might be a promising strategy to remediate dysbiosis.
Collapse
Affiliation(s)
- Anaïs B. Larabi
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, USA
| | - Hugo L. P. Masson
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, USA
| | - Andreas J. Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, USA
| |
Collapse
|
13
|
Abstract
Changes in the composition of the gut microbiota are associated with many human diseases. So far, however, we have failed to define homeostasis or dysbiosis by the presence or absence of specific microbial species. The composition and function of the adult gut microbiota is governed by diet and host factors that regulate and direct microbial growth. The host delivers oxygen and nitrate to the lumen of the small intestine, which selects for bacteria that use respiration for energy production. In the colon, by contrast, the host limits the availability of oxygen and nitrate, which results in a bacterial community that specializes in fermentation for growth. Although diet influences microbiota composition, a poor diet weakens host control mechanisms that regulate the microbiota. Hence, quantifying host parameters that control microbial growth could help define homeostasis or dysbiosis and could offer alternative strategies to remediate dysbiosis.
Collapse
Affiliation(s)
- Jee-Yon Lee
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA 95616, USA
| | - Renée M Tsolis
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA 95616, USA
| | - Andreas J Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA 95616, USA
| |
Collapse
|
14
|
Cheng X, Pi Z, Zheng Z, Liu S, Song F, Liu Z. Combined 16S rRNA gene sequencing and metabolomics to investigate the protective effects of Wu-tou Decoction on rheumatoid arthritis in rats. J Chromatogr B Analyt Technol Biomed Life Sci 2022; 1199:123249. [DOI: 10.1016/j.jchromb.2022.123249] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/06/2022] [Accepted: 04/07/2022] [Indexed: 12/12/2022]
|
15
|
Abstract
Abstract
Systemic sclerosis (SSc) is a rare autoimmune disease characterized by widespread microvasculopathy, inflammation, and fibrosis of the skin and internal organs. The involvement of the gastrointestinal tract is associated with a wide variety of symptoms and affects circa 90% of patients during the course of the disease. The gastrointestinal microbiota contains trillions of microbial cells and has been found to contribute to both local and systemic homeostasis. In both health and disease, a dynamic interrelationship between gut microbiome activity and the host immune system has been identified. Gastrointestinal dysbiosis has been described as having an important role in obesity, diabetes mellitus, liver disease, cardiovascular and neuropsychiatric disorders, neoplasia, as well as autoimmunity. Recent scientific data indicates a notable role of dysbiosis in the pathogenesis of SSc-related digestive involvement together with various other clinical manifestations. The present review aims to summarize the recent findings regarding digestive dysbiosis as well as the relationship between gastrointestinal microbiota and certain features of SSc.
Collapse
|
16
|
Zhang L, Qing P, Yang H, Wu Y, Liu Y, Luo Y. Gut Microbiome and Metabolites in Systemic Lupus Erythematosus: Link, Mechanisms and Intervention. Front Immunol 2021; 12:686501. [PMID: 34335588 PMCID: PMC8319742 DOI: 10.3389/fimmu.2021.686501] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/21/2021] [Indexed: 02/05/2023] Open
Abstract
Systemic lupus erythematosus (SLE), often considered the prototype of autoimmune diseases, is characterized by over-activation of the autoimmune system with abnormal functions of innate and adaptive immune cells and the production of a large number of autoantibodies against nuclear components. Given the highly complex and heterogeneous nature of SLE, the pathogenesis of this disease remains incompletely understood and is presumed to involve both genetic and environmental factors. Currently, disturbance of the gut microbiota has emerged as a novel player involved in the pathogenesis of SLE. With in-depth research, the understanding of the intestinal bacteria-host interaction in SLE is much more comprehensive. Recent years have also seen an increase in metabolomics studies in SLE with the attempt to identify potential biomarkers for diagnosis or disease activity monitoring. An intricate relationship between gut microbiome changes and metabolic alterations could help explain the mechanisms by which gut bacteria play roles in the pathogenesis of SLE. Here, we review the role of microbiota dysbiosis in the aetiology of SLE and how intestinal microbiota interact with the host metabolism axis. A proposed treatment strategy for SLE based on gut microbiome (GM) regulation is also discussed in this review. Increasing our understanding of gut microbiota and their function in lupus will provide us with novel opportunities to develop effective and precise diagnostic strategies and to explore potential microbiota-based treatments for patients with lupus.
Collapse
Affiliation(s)
- Lingshu Zhang
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Pingying Qing
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Hang Yang
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yongkang Wu
- Department of Laboratory Medicine and Outpatient, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Liu
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yubin Luo
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
17
|
Torun A, Hupalowska A, Trzonkowski P, Kierkus J, Pyrzynska B. Intestinal Microbiota in Common Chronic Inflammatory Disorders Affecting Children. Front Immunol 2021; 12:642166. [PMID: 34163468 PMCID: PMC8215716 DOI: 10.3389/fimmu.2021.642166] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/24/2021] [Indexed: 12/12/2022] Open
Abstract
The incidence and prevalence rate of chronic inflammatory disorders is on the rise in the pediatric population. Recent research indicates the crucial role of interactions between the altered intestinal microbiome and the immune system in the pathogenesis of several chronic inflammatory disorders in children, such as inflammatory bowel disease (IBD) and autoimmune diseases, such as type 1 diabetes mellitus (T1DM) and celiac disease (CeD). Here, we review recent knowledge concerning the pathogenic mechanisms underlying these disorders, and summarize the facts suggesting that the initiation and progression of IBD, T1DM, and CeD can be partially attributed to disturbances in the patterns of composition and abundance of the gut microbiota. The standard available therapies for chronic inflammatory disorders in children largely aim to treat symptoms. Although constant efforts are being made to maximize the quality of life for children in the long-term, sustained improvements are still difficult to achieve. Additional challenges are the changing physiology associated with growth and development of children, a population that is particularly susceptible to medication-related adverse effects. In this review, we explore new promising therapeutic approaches aimed at modulation of either gut microbiota or the activity of the immune system to induce a long-lasting remission of chronic inflammatory disorders. Recent preclinical studies and clinical trials have evaluated new approaches, for instance the adoptive transfer of immune cells, with genetically engineered regulatory T cells expressing antigen-specific chimeric antigen receptors. These approaches have revolutionized cancer treatments and have the potential for the protection of high-risk children from developing autoimmune diseases and effective management of inflammatory disorders. The review also focuses on the findings of studies that indicate that the responses to a variety of immunotherapies can be enhanced by strategic manipulation of gut microbiota, thus emphasizing on the importance of proper interaction between the gut microbiota and immune system for sustained health benefits and improvement of the quality of life of pediatric patients.
Collapse
Affiliation(s)
- Anna Torun
- Chair and Department of Biochemistry, Medical University of Warsaw, Warsaw, Poland
| | - Anna Hupalowska
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Piotr Trzonkowski
- Department of Medical Immunology, Medical University of Gdansk, Gdansk, Poland
| | - Jaroslaw Kierkus
- Department of Gastroenterology, Hepatology, Feeding Disorders and Pediatrics, The Children's Memorial Health Institute, Warsaw, Poland
| | - Beata Pyrzynska
- Chair and Department of Biochemistry, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
18
|
Sun P, Yang J, Wang B, Ma H, Zhang Y, Guo J, Chen X, Zhao J, Sun H, Yang J, Yang H, Cui Y. The effects of combined environmental factors on the intestinal flora of mice based on ground simulation experiments. Sci Rep 2021; 11:11373. [PMID: 34059794 PMCID: PMC8166921 DOI: 10.1038/s41598-021-91077-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 05/17/2021] [Indexed: 11/09/2022] Open
Abstract
The composition and function of intestinal microbial communities are important for human health. However, these intestinal floras are sensitive to changes in the environment. Adverse changes to intestinal flora can affect the health of astronauts, resulting in difficulties in implementing space missions. We randomly divided mice into three groups and placed each group in either a normal environment, simulated microgravity environment or a combined effects environment, which included simulated microgravity, low pressure and noise. Fecal samples of the mice were collected for follow-up analysis based on metagenomics technology. With the influence of different space environmental factors, the species composition at the phylum and genus levels were significantly affected by the combined effects environment, especially the abundance of the Firmicutes and Bacteroidetes. Furthermore, screening was conducted to identify biomarkers that could be regarded as environmental markers. And there have also been some noticeable changes in the function of intestinal floras. Moreover, the abundance of antibiotic resistance genes (ARGs) was also found to be changed under different environmental conditions, such as bacitracin and vancomycin. The combined effects environment could significantly affect the species composition, function, and the expression of ARGs of intestinal flora of mice which may provide a theoretical basis for space medical supervision and healthcare.
Collapse
Affiliation(s)
- Peiming Sun
- Department of General Surgery, Strategic Support Force Medical Center, Chaoyang District, Beijing, 100101, China
| | - Jiaqi Yang
- Department of General Surgery, Strategic Support Force Medical Center, Chaoyang District, Beijing, 100101, China
- Department of General Surgery, The 306th Hospital of Chinese PLA-Peking University Teaching Hospital, Chaoyang District, Beijing, 100101, China
| | - Bo Wang
- China Astronaut Research and Training Center, Haidian District, Beijing, 100094, China
| | - Huan Ma
- Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Haizhu District, Guangzhou, 510006, China
| | - Yin Zhang
- Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Haizhu District, Guangzhou, 510006, China
| | - Jinhu Guo
- Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Haizhu District, Guangzhou, 510006, China
| | - Xiaoping Chen
- China Astronaut Research and Training Center, Haidian District, Beijing, 100094, China
| | - Jianwei Zhao
- China Astronaut Research and Training Center, Haidian District, Beijing, 100094, China
| | - Hongwei Sun
- Department of General Surgery, Strategic Support Force Medical Center, Chaoyang District, Beijing, 100101, China
| | - Jianwu Yang
- Department of General Surgery, Strategic Support Force Medical Center, Chaoyang District, Beijing, 100101, China
| | - Heming Yang
- Department of General Surgery, Strategic Support Force Medical Center, Chaoyang District, Beijing, 100101, China.
| | - Yan Cui
- Department of General Surgery, Strategic Support Force Medical Center, Chaoyang District, Beijing, 100101, China.
| |
Collapse
|
19
|
Murdaca G, Greco M, Borro M, Gangemi S. Hygiene hypothesis and autoimmune diseases: A narrative review of clinical evidences and mechanisms. Autoimmun Rev 2021; 20:102845. [PMID: 33971339 DOI: 10.1016/j.autrev.2021.102845] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 02/20/2021] [Accepted: 02/27/2021] [Indexed: 12/12/2022]
Abstract
Since the start of the "modern era", characterized by the increase in urbanization, a progressive attention to hygiene and autoimmune conditions has considerably grown. Although these diseases are often multifactorial, it was demonstrated that environment factors such as pollution, diet and lifestyles may play a crucial role together with genetic signature. Our research, based on the newest and most significant literature of this topic, highlights that the progressive depletion of microbes and parasites due to increased socioeconomic improvement, may lead to a derangement of immunoregulatory mechanisms. Moreover, special attention was given to the complex interplay between microbial agents, as gut microbiome, diet and vitamin D supplementation with the aim of identifying promising future therapeutic options. In conclusion, autoimmunity cannot be limited to hygiene-hypothesis, but from the point of view of precision medicine, this theory represents a fundamental element together with the study of genomics, the microbiome and proteomics, in order to understand the complex functioning of the immune system.
Collapse
Affiliation(s)
- Giuseppe Murdaca
- Department of Internal Medicine, University of Genoa and IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Monica Greco
- Internal Medicine Department, San Paolo Hospital, 17100 Savona, Italy
| | - Matteo Borro
- Internal Medicine Department, San Paolo Hospital, 17100 Savona, Italy
| | - Sebastiano Gangemi
- School and Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| |
Collapse
|
20
|
Wu X, Chen X, Lyu X, Zheng H. Advances in Microbiome Detection Technologies and Application in Antirheumatic Drug Design. Curr Pharm Des 2021; 27:891-899. [PMID: 33308114 DOI: 10.2174/1381612826666201211114609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/25/2020] [Indexed: 11/22/2022]
Abstract
Rheumatic diseases are a kind of chronic inflammatory and autoimmune disease affecting the connection or supporting structures of the human body, such as the most common diseases Ankylosing spondylitis (AS), gout and Systemic lupus erythematosus (SLE). Although the precise etiology and pathogenesis of the different types of rheumatic diseases remain mostly unknown, it is now commonly believed that these diseases are attributed to some complex interactions between genetics and environmental factors, especially the gut microbiome. Altered microbiome showed clinical improvement in disease symptoms and partially restored to normality after prescribing disease-modifying antirheumatic drugs (DMARDs) or other treatment strategies. Recent advances in next-generation sequencing-based microbial profiling technology, especially metagenomics, have identified alteration of the composition and function of the gut microbiota in patients. Clinical and experimental data suggest that dysbiosis may play a pivotal role in the pathogenesis of these diseases. In this paper, we provide a brief review of the advances in the microbial profiling technology and up-to-date resources for accurate taxonomic assignment of metagenomic reads, which is a key step for metagenomics studies. In addition, we review the altered gut microbiota signatures that have been reported so far across various studies, upon which diagnostics classification models can be constructed, and the drug-induced regulation of the host microbiota can be used to control disease progression and symptoms.
Collapse
Affiliation(s)
- Xin Wu
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, China
| | - Xiang Chen
- Department of Bioinformatics, Hangzhou Nuowei Information Technology, Co., Ltd. Hangzhou, China
| | - Xiaochen Lyu
- Department of Bioinformatics, Hangzhou Nuowei Information Technology, Co., Ltd. Hangzhou, China
| | - Hao Zheng
- Department of Bioinformatics, Hangzhou Nuowei Information Technology, Co., Ltd. Hangzhou, China
| |
Collapse
|
21
|
Gut microbiota-microRNA interactions in ankylosing spondylitis. Autoimmun Rev 2021; 20:102827. [PMID: 33864943 DOI: 10.1016/j.autrev.2021.102827] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 02/17/2021] [Indexed: 12/20/2022]
Abstract
Ankylosing spondylitis (AS) is a chronic autoimmune inflammatory disability that is part of the rheumatic disease group of spondyloarthropathies. AS commonly influences the joints of the axial skeleton. The contributions to AS pathogenesis of genetic susceptibility (particularly HLA-B27 and ERAP-1) and epigenetic modifications, like non-coding RNAs, as well as environmental factors, have been investigated over the last few years. But the fundamental etiology of AS remains elusive to date. The evidence summarized here indicates that in the immunopathogenesis of AS, microRNAs and the gut microbiome perform critical functions. We discuss significant advances in the immunological mechanisms underlying AS and address potential cross-talk between the gut microbiome and host microRNAs. This critical interaction implicates a co-evolutionary symbiotic link between host immunity and the gut microbiome.
Collapse
|
22
|
Verburg I, van Veelen HPJ, Waar K, Rossen JWA, Friedrich AW, Hernández Leal L, García-Cobos S, Schmitt H. Effects of Clinical Wastewater on the Bacterial Community Structure from Sewage to the Environment. Microorganisms 2021; 9:718. [PMID: 33807193 PMCID: PMC8065902 DOI: 10.3390/microorganisms9040718] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/21/2021] [Accepted: 03/26/2021] [Indexed: 12/30/2022] Open
Abstract
This study pertains to measure differences in bacterial communities along the wastewater pathway, from sewage sources through the environment. Our main focus was on taxa which include pathogenic genera, and genera harboring antibiotic resistance (henceforth referred to as "target taxa"). Our objective was to measure the relative abundance of these taxa in clinical wastewaters compared to non-clinical wastewaters, and to investigate what changes can be detected along the wastewater pathway. The study entailed a monthly sampling campaign along a wastewater pathway, and taxa identification through 16S rRNA amplicon sequencing. Results indicated that clinical and non-clinical wastewaters differed in their overall bacterial composition, but that target taxa were not enriched in clinical wastewater. This suggests that treatment of clinical wastewater before release into the wastewater system would only remove a minor part of the potential total pathogen load in wastewater treatment plants. Additional findings were that the relative abundance of most target taxa was decreased after wastewater treatment, yet all investigated taxa were detected in 68% of the treated effluent samples-meaning that these bacteria are continuously released into the receiving surface water. Temporal variation was only observed for specific taxa in surface water, but not in wastewater samples.
Collapse
Affiliation(s)
- Ilse Verburg
- Wetsus, European Centre of Excellence for Sustainable Water Technology, 8900 CC Leeuwarden, The Netherlands; (I.V.); (H.P.J.v.V.); (L.H.L.)
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (J.W.A.R.); (A.W.F.); (S.G.-C.)
| | - H. Pieter J. van Veelen
- Wetsus, European Centre of Excellence for Sustainable Water Technology, 8900 CC Leeuwarden, The Netherlands; (I.V.); (H.P.J.v.V.); (L.H.L.)
| | - Karola Waar
- Izore, Centrum Infectieziekten Friesland, 8900 JA Leeuwarden, The Netherlands;
| | - John W. A. Rossen
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (J.W.A.R.); (A.W.F.); (S.G.-C.)
| | - Alex W. Friedrich
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (J.W.A.R.); (A.W.F.); (S.G.-C.)
| | - Lucia Hernández Leal
- Wetsus, European Centre of Excellence for Sustainable Water Technology, 8900 CC Leeuwarden, The Netherlands; (I.V.); (H.P.J.v.V.); (L.H.L.)
| | - Silvia García-Cobos
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (J.W.A.R.); (A.W.F.); (S.G.-C.)
| | - Heike Schmitt
- Wetsus, European Centre of Excellence for Sustainable Water Technology, 8900 CC Leeuwarden, The Netherlands; (I.V.); (H.P.J.v.V.); (L.H.L.)
- Institute for Risk Assessment Sciences, Utrecht University, 3508 TD Utrecht, The Netherlands
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), 3721 MA Bilthoven, The Netherlands
| |
Collapse
|
23
|
Sikora M, Stec A, Chrabaszcz M, Giebultowicz J, Samborowska E, Jazwiec R, Dadlez M, Olszewska M, Rudnicka L. Clinical Implications of Intestinal Barrier Damage in Psoriasis. J Inflamm Res 2021; 14:237-243. [PMID: 33542642 PMCID: PMC7851376 DOI: 10.2147/jir.s292544] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 12/24/2020] [Indexed: 12/12/2022] Open
Abstract
Background An increasing amount of evidence suggests an association between increased intestinal permeability and the pathogenesis of chronic inflammatory diseases. However, the clinical significance of gut barrier dysfunction in psoriasis remains to be established. Objective To evaluate whether there are differences in disease activity, the severity of gastrointestinal symptoms and the blood concentration of bacterial metabolites in psoriatic patients with a normal and altered intestinal barrier. Patients and Methods Gut barrier integrity was assessed with the serum concentrations of claudin-3, a modulator of intestinal tight junctions and an intestinal fatty acid-binding protein, a marker of enterocyte damage. Gastrointestinal symptoms were evaluated with a validated questionnaire. The concentration of trimethylamine N-oxide (TMAO), a gut microbiota-associated metabolite, was measured with high-performance liquid chromatography. Results One hundred and fourteen patients with psoriasis were finally enrolled in the study – 68 with an altered gut barrier and 46 with a properly functioning intestinal barrier. Patients with an altered gut barrier showed a significantly higher score in the Gastrointestinal Symptom Rating Scale (3.20 vs 1.46, p<0.001). Moreover, patients with psoriasis and a disrupted intestinal barrier demonstrated a higher disease activity (PASI: 19.7 vs 10.3, p<0.001) and systemic inflammatory parameters (neutrophil-to-lymphocyte ratio: 2.86 vs 1.71, p<0.001; C-reactive protein 3.76 vs 1.92; p<0.05). The marker of bacterial translocation was significantly higher in psoriatic patients with damaged gut integrity (TMAO: 375.7±51.9 vs 119.4±27.5 ng/mL; p<0.05). Conclusion The altered gut barrier in psoriasis is associated with gastrointestinal symptoms, systemic inflammatory profile and the increased blood concentration of gut microbiota-derived metabolite – TMAO. Intestinal barrier modulation represents a new promising therapeutic approach.
Collapse
Affiliation(s)
- Mariusz Sikora
- Department of Dermatology, Medical University of Warsaw, Warsaw, Poland
| | - Albert Stec
- Department of Dermatology, Medical University of Warsaw, Warsaw, Poland
| | | | - Joanna Giebultowicz
- Department of Bioanalysis and Drug Analysis, Faculty of Pharmacy with the Laboratory Medicine Division, Medical University of Warsaw, Warsaw, Poland
| | - Emilia Samborowska
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Radoslaw Jazwiec
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Michal Dadlez
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland.,Institute of Genetics and Biotechnology, Biology Department, Warsaw University, Warsaw, Poland
| | | | - Lidia Rudnicka
- Department of Dermatology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
24
|
Dooley LM, Ahmad TB, Pandey M, Good MF, Kotiw M. Rheumatic heart disease: A review of the current status of global research activity. Autoimmun Rev 2020; 20:102740. [PMID: 33333234 DOI: 10.1016/j.autrev.2020.102740] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 09/04/2020] [Indexed: 01/17/2023]
Abstract
Rheumatic heart disease (RHD) is a serious and long-term consequence of acute rheumatic fever (ARF), an autoimmune sequela of a mucosal infection by Streptococcus pyogenes (Group A Streptococcus, Strep A). The pathogenesis of ARF and RHD is complex and not fully understood but involves host and bacterial factors, molecular mimicry, and aberrant host innate and adaptive immune responses that result in loss of self-tolerance and subsequent cross-reactivity with host tissues. RHD is entirely preventable yet claims an estimated 320 000 lives annually. The major burden of disease is carried by developing nations and Indigenous populations within developed nations, including Australia. This review will focus on the epidemiology, pathogenesis and treatment of ARF and RHD in Australia, where: streptococcal pyoderma, rather than streptococcal pharyngitis, and Group C and Group G Streptococcus, have been implicated as antecedents to ARF; the rates of RHD in remote Indigenous communities are persistently among the highest in the world; government register-based programs coordinate disease screening and delivery of prophylaxis with variable success; and researchers are making significant progress in the development of a broad-spectrum vaccine against Strep A.
Collapse
Affiliation(s)
- Leanne M Dooley
- School of Health and Wellbeing, University of Southern Queensland, Toowoomba, Queensland, Australia; Institute for Life Sciences and the Environment, University of Southern Queensland, Toowoomba, Queensland, Australia.
| | - Tarek B Ahmad
- School of Health and Wellbeing, University of Southern Queensland, Toowoomba, Queensland, Australia; Institute for Life Sciences and the Environment, University of Southern Queensland, Toowoomba, Queensland, Australia.
| | - Manisha Pandey
- The Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia.
| | - Michael F Good
- The Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia.
| | - Michael Kotiw
- School of Health and Wellbeing, University of Southern Queensland, Toowoomba, Queensland, Australia; Institute for Life Sciences and the Environment, University of Southern Queensland, Toowoomba, Queensland, Australia.
| |
Collapse
|
25
|
Amino Acid Metabolism in Rheumatoid Arthritis: Friend or Foe? Biomolecules 2020; 10:biom10091280. [PMID: 32899743 PMCID: PMC7563518 DOI: 10.3390/biom10091280] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 02/07/2023] Open
Abstract
In mammals, amino acid metabolism has evolved to act as a critical regulator of innate and adaptive immune responses. Rheumatoid arthritis (RA) is the most common form of inflammatory arthropathy sustained by autoimmune responses. We examine here the current knowledge of tryptophan and arginine metabolisms and the main immunoregulatory pathways in amino acid catabolism, in both RA patients and experimental models of arthritis. We found that l-tryptophan (Trp) metabolism and, in particular, the kynurenine pathway would exert protective effects in all experimental models and in some, but not all, RA patients, possibly due to single nucleotide polymorphisms in the gene coding for indoleamine 2,3-dioxygenase 1 (IDO1; the enzyme catalyzing the rate-limiting step of the kynurenine pathway). The function, i.e., either protective or pathogenetic, of the l-arginine (Arg) metabolism in RA was less clear. In fact, although immunoregulatory arginase 1 (ARG1) was highly induced at the synovial level in RA patients, its true functional role is still unknown, possibly because of few available preclinical data. Therefore, our analysis would indicate that amino acid metabolism represents a fruitful area of research for new drug targets for a more effective and safe therapy of RA and that further studies are demanding to pursue such an important objective.
Collapse
|