1
|
Liu J, Li B, Zhou X, Liu G, Li C, Hu Z, Peng R. Uncovering the mechanisms of Zhubi decoction against rheumatoid arthritis through an integrated study of network pharmacology, metabolomics, and intestinal flora. JOURNAL OF ETHNOPHARMACOLOGY 2025; 336:118736. [PMID: 39186991 DOI: 10.1016/j.jep.2024.118736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 08/28/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zhubi Decoction (ZBD) is a modified formulation derived from the classic traditional Chinese medicine prescription "Er-Xian Decoction" documented in the esteemed "Clinical Manual of Chinese Medical Prescription". While the utilization of ZBD has exhibited promising clinical outcomes in treating rheumatoid arthritis (RA), the precise bioactive chemical constituents and the underlying mechanisms involved in its therapeutic efficacy remain to be comprehensively determined. AIM OF THE STUDY This study aims to systematically examine ZBD's pharmacological effects and molecular mechanisms for RA alleviation. MATERIALS AND METHODS Utilizing the collagen-induced arthritis (CIA) rat model, we comprehensively evaluated the anti-rheumatoid arthritis effects of ZBD in vivo through various indices, such as paw edema, arthritis index, ankle diameter, inflammatory cytokine levels, pathological conditions, and micro-CT analysis. The UPLC-MS/MS technique was utilized to analyze the compounds of ZBD. The potential therapeutic targets and signaling pathways of ZBD in the management of RA were predicted using network pharmacology. To analyze comprehensive metabolic profiles and identify underlying metabolic pathways, we conducted a serum-based widely targeted metabolomics analysis utilizing LC-MS technology. Key targets and predicted pathways were further validated using immunofluorescent staining, which integrated findings from serum metabolomics and network pharmacology analysis. Additionally, we analyzed the gut microbiota composition in rats employing 16 S rDNA sequencing and investigated the effects of ZBD on the microbiota of CIA rats through bioinformatics and statistical methods. RESULTS ZBD exhibited remarkable efficacy in alleviating RA symptoms in CIA rats without notable side effects. This included reduced paw redness and swelling, minimized joint damage, improved the histopathology of cartilage and synovium, mitigated the inflammatory state, and lowered serum concentrations of cytokines TNF-α, IL-1β and IL-6. Notably, the effectiveness of ZBD was comparable to MTX. Network pharmacology analysis revealed inflammation and immunity-related signaling pathways, such as PI3K/AKT, MAPK, IL-17, and TNF signaling pathways, as vital mediators in the effectual mechanisms of ZBD. Immunofluorescence analysis validated ZBD's ability to inhibit PI3K/AKT pathway proteins. Serum metabolomics studies revealed that ZBD modulates 170 differential metabolites, partially restored disrupted metabolic profiles in CIA rats. With a notable impact on amino acids and their metabolites, and lipids and lipid-like molecules. Integrated analysis of metabolomics and network pharmacology identified 6 pivotal metabolite pathways and 3 crucial targets: PTGS2, GSTP1, and ALDH2. Additionally, 16 S rDNA sequencing illuminated that ZBD mitigated gut microbiota dysbiosis in the CIA group, highlighting key genera such as Ligilactobacillus, Prevotella_9, unclassified_Bacilli, and unclassified_rumen_bacterium_JW32. Correlation analysis disclosed a significant link between 47 distinct metabolites and specific bacterial species. CONCLUSION ZBD is a safe and efficacious TCM formulation, demonstrates efficacy in treating RA through its multi-component, multi-target, and multi-pathway mechanisms. The regulation of inflammation and immunity-related signaling pathways constitutes a crucial mechanism of ZBD's efficacy. Furthermore, ZBD modulates host metabolism and intestinal flora. The integrated analysis presents experimental evidence of ZBD for the management of RA.
Collapse
Affiliation(s)
- Jing Liu
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430061, China.
| | - Bocun Li
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430061, China.
| | - Xiaohong Zhou
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430061, China.
| | - Guangya Liu
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430061, China.
| | - Chao Li
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430061, China.
| | - Zhaoduan Hu
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430061, China.
| | - Rui Peng
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430061, China.
| |
Collapse
|
2
|
Kunath BJ, De Rudder C, Laczny CC, Letellier E, Wilmes P. The oral-gut microbiome axis in health and disease. Nat Rev Microbiol 2024; 22:791-805. [PMID: 39039286 DOI: 10.1038/s41579-024-01075-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2024] [Indexed: 07/24/2024]
Abstract
The human body hosts trillions of microorganisms throughout many diverse habitats with different physico-chemical characteristics. Among them, the oral cavity and the gut harbour some of the most dense and diverse microbial communities. Although these two sites are physiologically distinct, they are directly connected and can influence each other in several ways. For example, oral microorganisms can reach and colonize the gastrointestinal tract, particularly in the context of gut dysbiosis. However, the mechanisms of colonization and the role that the oral microbiome plays in causing or exacerbating diseases in other organs have not yet been fully elucidated. Here, we describe recent advances in our understanding of how the oral and intestinal microbiota interplay in relation to their impact on human health and disease.
Collapse
Affiliation(s)
- Benoit J Kunath
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
| | - Charlotte De Rudder
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Cedric C Laczny
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Elisabeth Letellier
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Paul Wilmes
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg.
| |
Collapse
|
3
|
Kim HJ, Park DH, Han SH, Kim SY. Optimal storage time and temperature of human oral samples to minimize microbiome changes: A scoping review. JAPANESE DENTAL SCIENCE REVIEW 2024; 60:220-231. [PMID: 39498230 PMCID: PMC11533461 DOI: 10.1016/j.jdsr.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 03/25/2024] [Accepted: 05/12/2024] [Indexed: 11/07/2024] Open
Abstract
Improper storage times and temperatures negatively impact the quality of biospecimens with oral microbiomes. This study aimed to determine the optimal storage time and temperature for maintaining the integrity of human dental plaque and saliva samples' microbial profiles. A comprehensive search yielded 5433 studies, with 12 meeting inclusion criteria. The number of studies on the storage time and temperature for plaque or saliva samples was extremely limited, with large variability in study designs and analytical tools. The best approach for dental plaque and saliva samples was to immediately freeze fresh specimens at - 80 °C or lower until DNA extraction, with a recommended storage time not exceeding 1-2 years, regardless of temperature. Checkerboard DNA-DNA hybridization-based studies suggested dental plaque storage at - 20 °C for 6 months, but a shorter duration was advised. Based on 16 S rRNA gene sequencing studies, dental plaque samples could be stored at - 80 °C for 6 months in 75 % ethanol or Bead Solution. Dental plaque and saliva samples could be stored at room temperature for 1-2 weeks without significant microbiome changes if stored in appropriate media. Further well-designed randomized controlled studies with longer-storage duration are necessary to establish more definitive guidelines.
Collapse
Affiliation(s)
- Hyun Ju Kim
- Department of Periodontics, Seoul National University Dental Hospital, 101 Daehakro, Jongno-gu, Seoul 03080, Republic of Korea
- Department of Periodontology and Dental Research Institute, School of Dentistry, Seoul National University, 101 Daehakro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Dong Hyun Park
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Sun-Young Kim
- Department of Conservative Dentistry and Dental Research Institute, School of Dentistry, Seoul National University, 101 Daehakro, Jongno-gu, Seoul 03080, Republic of Korea
| |
Collapse
|
4
|
Kuo CL, Hsin-Hsien Yeh S, Chang TM, I-Chin Wei A, Chen WJ, Chu HF, Tseng AL, Lin PY, Lin ZC, Peng KT, Liu JF. Bacillus coagulans BACO-17 ameliorates in vitro and in vivo progression of Rheumatoid arthritis. Int Immunopharmacol 2024; 141:112863. [PMID: 39146779 DOI: 10.1016/j.intimp.2024.112863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 07/31/2024] [Accepted: 07/31/2024] [Indexed: 08/17/2024]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that causes persistent inflammation involving the joints, cartilage, and synovium. In individuals with RA, alterations in the composition of intestinal bacteria suggest the vital role of gut microbiota in immune dysfunction. Multiple therapies commonly used to treat RA can also alter the diversity of gut microbiota, further suggesting the modulation of gut microbiota as a prevention or treatment for RA. Therefore, a better understanding of the changes in the gut microbiota that accompany RA should facilitate the development of novel therapeutic approaches. In this study, B. coagulans BACO-17 not only significantly reduced paw swelling, arthritis scores, and hind paw and forepaw thicknesses but also protected articular cartilage and the synovium against RA degeneration, with a corresponding downregulation of TNF-α expression. The inhibition or even reversing of RA progression highlights B. coagulans BACO-17 as a novel therapeutic for RA worth investigating.
Collapse
Affiliation(s)
- Chun-Lin Kuo
- Department of Surgery, Tri-Service General Hospital Keelung Branch, Keelung, Taiwan; Department of Orthopedic Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Skye Hsin-Hsien Yeh
- School of Medicine, National Defense Medical Center, Taipei, Taiwan; Brain Research Center, National Yang Ming Chiao Tung University, Taipei City, Taiwan
| | - Tsung-Ming Chang
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei City, Taiwan
| | - Augusta I-Chin Wei
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wei-Jen Chen
- Research and Development Department, Syngen Biotech Co., Ltd., Tainan, Taiwan; Graduate Institute of Management, Minghsin University of Science and Technology, Hsinchu, Taiwan
| | - Hui-Fang Chu
- Research and Development Department, Syngen Biotech Co., Ltd., Tainan, Taiwan
| | - Ai-Lun Tseng
- Research and Development Department, Syngen Biotech Co., Ltd., Tainan, Taiwan
| | - Pai-Yin Lin
- Research and Development Department, Syngen Biotech Co., Ltd., Tainan, Taiwan
| | - Zih-Chan Lin
- Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi City, Chiayi County, Taiwan
| | - Kuo-Ti Peng
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Puzi City, Chiayi County, Taiwan.
| | - Ju-Fang Liu
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan; Translational Medicine Center, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei City, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.
| |
Collapse
|
5
|
Ji Y, Sun H, Wang Y, Li Y, Piao R, Bu L, Xu H. Characterizing the oral and gastrointestinal microbiome associated with healthy aging: insights from long-lived populations in Northeastern China. GeroScience 2024:10.1007/s11357-024-01419-2. [PMID: 39505797 DOI: 10.1007/s11357-024-01419-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 10/29/2024] [Indexed: 11/08/2024] Open
Abstract
The oral and gastrointestinal (GI) tract microbiota in humans is susceptible to geographical influences and represents vital factors impacting healthy aging. The northeastern region of China, characterized by distinct dietary and climatic conditions, significantly influences the human microbiome composition. However, the microbial structure of the entire long-lived population in this area has not been evaluated. This study recruited a cohort of 142 individuals aged 55-102 residing in Northeast China, and their oral and gut microbiota were evaluated using full-length 16S rRNA gene amplicon sequencing. The results indicate that the oral and GI tract microbiota of long-lived individuals showed reduced microbial taxonomic richness and evenness compared to sub-longevity individuals. With aging, the core species experience a gradual decline in abundance, while subordinate species show an increase. The long-lived population exhibited a heightened ability to enrich beneficial bacteria including Akkermansia, Alistipes, Parabacteroides, and Eubacterium coprostanoligenes in the GI tract, which are associated with host metabolism and have the potential to act as probiotics, reducing the risks of unhealthy aging in the northeast population. Bifidobacterium sp. and Lactobacillus salivarius have been found to coexist in both the oral cavity and the GI tract of long-lived individuals. We hypothesize that beneficial bacterial taxa from the oral cavity colonize the GI tract more extensively in long-lived individuals compared to those with a shorter lifespan. These findings pave the way for identifying probiotic strains that can promote healthy aging in Northeast China.
Collapse
Affiliation(s)
- Yue Ji
- Jinqiu Hospital of Liaoning Province, Shenyang, 110016, China
| | - Hao Sun
- CAS Key Laboratory of Forest Ecology and Management, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang, 110016, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Yingda Wang
- Jinqiu Hospital of Liaoning Province, Shenyang, 110016, China
| | - Yanhui Li
- Jinqiu Hospital of Liaoning Province, Shenyang, 110016, China
| | - Rennv Piao
- Jinqiu Hospital of Liaoning Province, Shenyang, 110016, China
| | - Li Bu
- Jinqiu Hospital of Liaoning Province, Shenyang, 110016, China.
| | - Hui Xu
- CAS Key Laboratory of Forest Ecology and Management, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang, 110016, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
6
|
Ma F, Li Z, Liu H, Chen S, Zheng S, Zhu J, Shi H, Ye H, Qiu Z, Gao L, Han B, Yang Q, Wang X, Zhang Y, Cheng L, Fan H, Lv S, Zhao X, Zhou H, Li J, Hong M. Dietary-timing-induced gut microbiota diurnal oscillations modulate inflammatory rhythms in rheumatoid arthritis. Cell Metab 2024; 36:2367-2382.e5. [PMID: 39260371 DOI: 10.1016/j.cmet.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/10/2024] [Accepted: 08/12/2024] [Indexed: 09/13/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune condition characterized by inflammatory activity with distinct rhythmic fluctuations. However, the precise mechanisms governing these inflammatory rhythms remain elusive. Here, we explore the interaction between dietary patterns, gut microbiota diurnal oscillations, and the rhythmicity of RA in both collagen-induced arthritis (CIA) mice and patients with RA and highlight the significance of dietary timing in modulating RA inflammatory rhythms linked to gut microbiota. Specifically, we discovered that Parabacteroides distasonis (P. distasonis) uses β-glucosidase (β-GC) to release glycitein (GLY) from the diet in response to daily nutritional cues, influencing RA inflammatory rhythms dependent on the sirtuin 5-nuclear factor-κB (SIRT5-NF-κB) axis. Notably, we validated the daily fluctuations of P. distasonis-β-GC-GLY in patients with RA through continuous sampling across day-night cycles. These findings underscore the crucial role of dietary timing in RA rhythmicity and propose potential clinical implications for novel therapeutic strategies to alleviate arthritis.
Collapse
Affiliation(s)
- Fopei Ma
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China; Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510000, China
| | - Zhuang Li
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China.
| | - Haihua Liu
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Shixian Chen
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Songyuan Zheng
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Junqing Zhu
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Hao Shi
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510000, China
| | - Haixin Ye
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510000, China
| | - Zhantu Qiu
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510000, China
| | - Lei Gao
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510000, China
| | - Bingqi Han
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510000, China
| | - Qian Yang
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Xing Wang
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Yang Zhang
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Lifang Cheng
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Huijie Fan
- Department of Traditional Chinese Medicine, People's Hospital of Yangjiang, Yangjiang 529500, China
| | - Shuaijun Lv
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xiaoshan Zhao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Hongwei Zhou
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China.
| | - Juan Li
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China; Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510000, China.
| | - Mukeng Hong
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China.
| |
Collapse
|
7
|
Ryu EP, Gautam Y, Proctor DM, Bhandari D, Tandukar S, Gupta M, Gautam GP, Relman DA, Shibl AA, Sherchand JB, Jha AR, Davenport ER. Nepali oral microbiomes reflect a gradient of lifestyles from traditional to industrialized. MICROBIOME 2024; 12:228. [PMID: 39497165 PMCID: PMC11533410 DOI: 10.1186/s40168-024-01941-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 09/27/2024] [Indexed: 11/06/2024]
Abstract
BACKGROUND Lifestyle plays an important role in shaping the gut microbiome. However, its contributions to the oral microbiome remain less clear, due to the confounding effects of geography and methodology in investigations of populations studied to date. Furthermore, while the oral microbiome seems to differ between foraging and industrialized populations, we lack insight into whether transitions to and away from agrarian lifestyles shape the oral microbiota. Given the growing interest in so-called "vanishing microbiomes" potentially being a risk factor for increased disease prevalence in industrialized populations, it is important that we distinguish lifestyle from geography in the study of microbiomes across populations. RESULTS Here, we investigate salivary microbiomes of 63 Nepali individuals representing a spectrum of lifestyles: foraging, subsistence farming (individuals that transitioned from foraging to farming within the last 50 years), agriculturalists (individuals that have transitioned to farming for at least 300 years), and industrialists (expatriates that immigrated to the USA within the last 20 years). We characterize the role of lifestyle in microbial diversity, identify microbes that differ between lifestyles, and pinpoint specific lifestyle factors that may be contributing to differences in the microbiomes across populations. Contrary to prevailing views, when geography is controlled for, oral microbiome alpha diversity does not differ significantly across lifestyles. Microbiome composition, however, follows the gradient of lifestyles from foraging through agrarianism to industrialism, supporting the notion that lifestyle indeed plays a role in the oral microbiome. Relative abundances of several individual taxa, including Streptobacillus and an unclassified Porphyromonadaceae genus, also mirror lifestyle. Finally, we identify specific lifestyle factors associated with microbiome composition across the gradient of lifestyles, including smoking and grain sources. CONCLUSION Our findings demonstrate that by studying populations within Nepal, we can isolate an important role of lifestyle in determining oral microbiome composition. In doing so, we highlight the potential contributions of several lifestyle factors, underlining the importance of carefully examining the oral microbiome across lifestyles to improve our understanding of global microbiomes. Video Abstract.
Collapse
Affiliation(s)
- Erica P Ryu
- Department of Biology, Pennsylvania State University, University Park, PA, USA
| | - Yoshina Gautam
- Genetic Heritage Group, Program in Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Diana M Proctor
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Dinesh Bhandari
- Public Health Research Laboratory, Institute of Medicine, Maharajgunj, Kathmandu, Nepal
- School of Public Health, University of Adelaide, Adelaide, SA, Australia
| | - Sarmila Tandukar
- Public Health Research Laboratory, Institute of Medicine, Maharajgunj, Kathmandu, Nepal
- Organization for Public Health and Environment Management, Lalitpur, Bagmati, Nepal
| | - Meera Gupta
- Department of Biology, Pennsylvania State University, University Park, PA, USA
- Sidney Kimmel Medical College, Philadelphia, PA, UAE
| | | | - David A Relman
- Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
- Section of Infectious Diseases, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Ahmed A Shibl
- Genetic Heritage Group, Program in Biology, New York University Abu Dhabi, Abu Dhabi, UAE
- Center for Genomics and Systems Biology, and Public Health Research Center, New York University Abu Dhabi, Abu Dhabi, UAE
| | | | - Aashish R Jha
- Genetic Heritage Group, Program in Biology, New York University Abu Dhabi, Abu Dhabi, UAE.
- Center for Genomics and Systems Biology, and Public Health Research Center, New York University Abu Dhabi, Abu Dhabi, UAE.
| | - Emily R Davenport
- Department of Biology, Pennsylvania State University, University Park, PA, USA.
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
8
|
Shimokawa C. The gut microbiome-helminth-immune axis in autoimmune diseases. Parasitol Int 2024; 104:102985. [PMID: 39491642 DOI: 10.1016/j.parint.2024.102985] [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: 09/26/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
The global prevalence of autoimmune diseases has surged in recent decades. Consequently, environmental triggers have emerged as crucial contributors to autoimmune diseases, equally relevant to classical risk factors, such as genetic polymorphisms, infections, and smoking. Sequencing-based approaches have demonstrated distinct gut microbiota compositions in individuals with autoimmune diseases, including multiple sclerosis, rheumatoid arthritis, type 1 diabetes mellitus (T1D), and systemic lupus erythematosus, compared to healthy controls. Furthermore, fecal microbiota transplantation and microbial inoculation experiments have supported the hypothesis that alterations in the gut microbiota can influence autoimmune responses and disease outcomes. Herein, we propose that intestinal helminths may serve as a critical factor in inducing alterations in the gut microbiota. The concept of helminth-mediated suppression of autoimmune diseases in humans is supported by substantial evidence, aligning with the long-standing "hygiene hypothesis." This review focused on T1D to explore the interactions between parasites, gut microbiota, and the immune system-a topic that remains a black box within this intricate triangular relationship.
Collapse
Affiliation(s)
- Chikako Shimokawa
- Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan.
| |
Collapse
|
9
|
Zhu X, Long W, Zhang J, Jian C, Chen J, Huang J, Li S, Zhang J, Wang L, Chen Y, Wu J, Wang T, Zou Q, Zhu J, Zeng F. Integrated multi-omics revealed that dysregulated lipid metabolism played an important role in RA patients with metabolic diseases. Arthritis Res Ther 2024; 26:188. [PMID: 39482717 PMCID: PMC11529425 DOI: 10.1186/s13075-024-03423-5] [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: 07/17/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024] Open
Abstract
OBJECTIVES Patients with rheumatoid arthritis (RA) commonly experience a high prevalence of multiple metabolic diseases (MD), leading to higher morbidity and premature mortality. Here, we aimed to investigate the pathogenesis of MD in RA patients (RA_MD) through an integrated multi-omics approach. METHODS Fecal and blood samples were collected from a total of 181 subjects in this study for multi-omics analyses, including 16S rRNA and internally transcribed spacer (ITS) gene sequencing, metabolomics, transcriptomics, proteomics and phosphoproteomics. Spearman's correlation and protein-protein interaction networks were used to assess the multi-omics data correlations. The Least Absolute Shrinkage and Selection Operator (LASSO) machine learning algorithm were used to identify disease-specific biomarkers for RA_MD diagnosis. RESULTS Our results found that RA_MD was associated with differential abundance of gut microbiota such as Turicibacter and Neocosmospora, metabolites including decreased unsaturated fatty acid, genes related to linoleic acid metabolism and arachidonic acid metabolism, as well as downregulation of proteins and phosphoproteins involved in cholesterol metabolism. Furthermore, a multi-omics classifier differentiated RA_MD from RA with high accuracy (AUC: 0.958). Compared to gouty arthritis and systemic lupus erythematosus, dysregulation of lipid metabolism showed disease-specificity in RA_MD. CONCLUSIONS The integration of multi-omics data demonstrates that lipid metabolic pathways play a crucial role in RA_MD, providing the basis and direction for the prevention and early diagnosis of MD, as well as new insights to complement clinical treatment options.
Collapse
Affiliation(s)
- Xiaoting Zhu
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Wubin Long
- Department of Rheumatology and Immunology, Sichuan Provincial People's Hospital, Chengdu, China
| | - Jing Zhang
- Department of Rheumatology and Immunology, Sichuan Provincial People's Hospital, Chengdu, China
| | - Congcong Jian
- School of Basic Medical Science, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianghua Chen
- School of Basic Medical Science, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jiaxin Huang
- School of Basic Medical Science, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Shilin Li
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Jie Zhang
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Liang Wang
- Department of Rheumatology and Immunology, Sichuan Provincial People's Hospital, Chengdu, China
| | - Yan Chen
- Department of Rheumatology and Immunology, Sichuan Provincial People's Hospital, Chengdu, China
| | - Jianhong Wu
- Department of Rheumatology and Immunology, Dazhou Central Hospital, Dazhou, China
| | - Tingting Wang
- Department of Rheumatology and Immunology, Dazhou Central Hospital, Dazhou, China
| | - Qinghua Zou
- Department of Rheumatology and Immunology, First Affiliated Hospital of Army Medical University, Chongqing, China.
| | - Jing Zhu
- Department of Rheumatology and Immunology, Sichuan Provincial People's Hospital, Chengdu, China.
| | - Fanxin Zeng
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan, China.
| |
Collapse
|
10
|
Takewaki D, Kiguchi Y, Masuoka H, Manu MS, Raveney BJE, Narushima S, Kurokawa R, Ogata Y, Kimura Y, Sato N, Ozawa Y, Yagishita S, Araki T, Miyake S, Sato W, Suda W, Yamamura T. Tyzzerella nexilis strains enriched in mobile genetic elements are involved in progressive multiple sclerosis. Cell Rep 2024; 43:114785. [PMID: 39341204 DOI: 10.1016/j.celrep.2024.114785] [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: 02/09/2024] [Revised: 08/19/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Multiple sclerosis (MS) is an autoimmune-demyelinating disease with an inflammatory pathology formed by self-reactive lymphocytes with activated glial cells. Progressive MS, characterized by resistance to medications, significantly differs from the non-progressive form in gut microbiome profiles. After confirming an increased abundance of "Tyzzerella nexilis" in various cohorts of progressive MS, we identified a distinct cluster of T. nexilis strains enriched in progressive MS based on long-read metagenomics. The distinct T. nexilis cluster is characterized by a large number of mobile genetic elements (MGEs) and a lack of defense systems against MGEs. Microbial genes for sulfate reduction and flagella formation with pathogenic implications are specific to this cluster. Moreover, these flagellar genes are encoded on MGEs. Mono-colonization with MGE-enriched T. nexilis made germ-free mice more susceptible to experimental autoimmune encephalomyelitis. These results indicate that the progression of MS may be promoted by MGE-enriched T. nexilis with potentially pathogenic properties.
Collapse
Affiliation(s)
- Daiki Takewaki
- Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan; Multiple Sclerosis Center, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan; Laboratory for Symbiotic Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Yuya Kiguchi
- Laboratory for Symbiotic Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8568, Japan
| | - Hiroaki Masuoka
- Laboratory for Symbiotic Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Mallahalli S Manu
- Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan; Multiple Sclerosis Center, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| | - Ben J E Raveney
- Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan; Multiple Sclerosis Center, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan; Laboratory for Symbiotic Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Seiko Narushima
- Laboratory for Mucosal Immunity, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Rina Kurokawa
- Laboratory for Symbiotic Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Yusuke Ogata
- Laboratory for Symbiotic Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Yukio Kimura
- Multiple Sclerosis Center, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan; Department of Radiology, National Center of Neurology and Psychiatry Hospital, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| | - Noriko Sato
- Multiple Sclerosis Center, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan; Department of Radiology, National Center of Neurology and Psychiatry Hospital, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| | - Yusuke Ozawa
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| | - Sosuke Yagishita
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| | - Toshiyuki Araki
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| | - Sachiko Miyake
- Department of Immunology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Wakiro Sato
- Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan; Multiple Sclerosis Center, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| | - Wataru Suda
- Laboratory for Symbiotic Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan.
| | - Takashi Yamamura
- Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan; Multiple Sclerosis Center, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan.
| |
Collapse
|
11
|
Catalan EA, Seguel-Fuentes E, Fuentes B, Aranguiz-Varela F, Castillo-Godoy DP, Rivera-Asin E, Bocaz E, Fuentes JA, Bravo D, Schinnerling K, Melo-Gonzalez F. Oral Pathobiont-Derived Outer Membrane Vesicles in the Oral-Gut Axis. Int J Mol Sci 2024; 25:11141. [PMID: 39456922 PMCID: PMC11508520 DOI: 10.3390/ijms252011141] [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: 09/06/2024] [Revised: 10/04/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Oral pathobionts are essential in instigating local inflammation within the oral cavity and contribute to the pathogenesis of diseases in the gastrointestinal tract and other distant organs. Among the Gram-negative pathobionts, Porphyromonas gingivalis and Fusobacterium nucleatum emerge as critical drivers of periodontitis, exerting their influence not only locally but also as inducers of gut dysbiosis, intestinal disturbances, and systemic ailments. This dual impact is facilitated by their ectopic colonization of the intestinal mucosa and the subsequent mediation of distal systemic effects by releasing outer membrane vesicles (OMVs) into circulation. This review elucidates the principal components of oral pathobiont-derived OMVs implicated in disease pathogenesis within the oral-gut axis, detailing virulence factors that OMVs carry and their interactions with host epithelial and immune cells, both in vitro and in vivo. Additionally, we shed light on the less acknowledged interplay between oral pathobionts and the gut commensal Akkermansia muciniphila, which can directly impede oral pathobionts' growth and modulate bacterial gene expression. Notably, OMVs derived from A. muciniphila emerge as promoters of anti-inflammatory effects within the gastrointestinal and distant tissues. Consequently, we explore the potential of A. muciniphila-derived OMVs to interact with oral pathobionts and prevent disease in the oral-gut axis.
Collapse
Affiliation(s)
- Eduardo A. Catalan
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Emilio Seguel-Fuentes
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Brandon Fuentes
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Felipe Aranguiz-Varela
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Daniela P. Castillo-Godoy
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Elizabeth Rivera-Asin
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Elisa Bocaz
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Juan A. Fuentes
- Laboratorio de Genética y Patogénesis Bacteriana, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile;
| | - Denisse Bravo
- Cellular Interactions Laboratory, Faculty of Dentistry, Universidad Andrés Bello, Santiago 8370133, Chile;
| | - Katina Schinnerling
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Felipe Melo-Gonzalez
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| |
Collapse
|
12
|
Song C, Liu F, Mei Y, Cai W, Cheng K, Guo D, Liu Y, Shi H, Duan DD, Liu Z. Integrated metagenomic and metabonomic mechanisms for the therapeutic effects of Duhuo Jisheng decoction on intervertebral disc degeneration. PLoS One 2024; 19:e0310014. [PMID: 39418241 PMCID: PMC11486403 DOI: 10.1371/journal.pone.0310014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 08/20/2024] [Indexed: 10/19/2024] Open
Abstract
Intervertebral disc degeneration (IVDD) is a prevalent orthopedic condition with lower back pain as the predominant clinical presentation that challenges clinical treatment with few therapeutic options. Duhuo Jisheng Decoction (DHJSD) has been proven effective in the therapy of IVDD, but the precise underlying mechanisms remain not fully elucidated. The current study was designed to test our hypothesis that DHJSD may systematically correct the phenotypic disruption of the gut microbiota and changes in the serum metabolome linked to IVDD. Analysis of the active ingredients of DHJSD by ultra high performance liquid chromatography. An integrated metagenomic and metabonomic approach was used to analyze feces and blood samples from normal and IVDD rats. Compared to the control group, fiber ring pinning on the caudal 3 to caudal 5 segments of the rats caused IVDD and significantly altered the compositions of the intestinal microbiota and serum metabolites. Integrated analysis revealed commonly-altered metabolic pathways shared by both intestinal microbiota and serum metabolome of the IVDD rats. DHJSD inhibited the degenerative process and restored the compositions of the perturbed gut microbiota, particularly the relative abundance of commensal microbes of the Prevotellaceae family. DHJSD also corrected the altered metabolic pathways involved in the metabolism of glycine, serine, threonine, valine, the citric acid cycle, and biosynthesis of leucine and isoleucine. DHJSD inhibited the disc degeneration process by an integrated metagenomic and metabonomic mechanism to restore the microbiome profile and normalize the metabonomic pathways.
Collapse
Affiliation(s)
- Chao Song
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Fei Liu
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Yongliang Mei
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Weiye Cai
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Kang Cheng
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Daru Guo
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Yong Liu
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Houyin Shi
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Dayue Darrel Duan
- Center for Phenomics of Traditional Chinese Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Zongchao Liu
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- Luzhou Longmatan District People’s Hospital, Luzhou, Sichuan Province, China
| |
Collapse
|
13
|
Cao Y, Chen J, Xiao J, Hong Y, Xu K, Zhu Y. Butyrate: a bridge between intestinal flora and rheumatoid arthritis. Front Immunol 2024; 15:1475529. [PMID: 39478858 PMCID: PMC11521846 DOI: 10.3389/fimmu.2024.1475529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 09/30/2024] [Indexed: 11/02/2024] Open
Abstract
In patients with rheumatoid arthritis (RA), intestinal flora imbalance and butyrate metabolism disorders precede clinical arthritis and are associated with the pathogenesis of RA. This imbalance can alter the immunology and intestinal permeability of the intestinal mucosa, leading to damage to the intestinal barrier. In this context, bacteria and their metabolites can enter the bloodstream and reach the distant target tissues of the host, resulting in local inflammation and aggravating arthritis. Additionally, arthritis is also exacerbated by bone destruction and immune tolerance due to disturbed differentiation of osteoclasts and adaptive immune cells. Of note, butyrate is a metabolite of intestinal flora, which not only locally inhibits intestinal immunity and targets zonulin and tight junction proteins to alleviate intestinal barrier-mediated arthritis but also inhibits osteoclasts and autoantibodies and balances the immune responses of T and B lymphocytes throughout the body to repress bone erosion and inflammation. Therefore, butyrate is a key intermediate linking intestinal flora to the host. As a result, restoring the butyrate-producing capacity of intestinal flora and using exogenous butyrate are potential therapeutic strategies for RA in the future.
Collapse
Affiliation(s)
- Yang Cao
- Second Clinical College, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Jingjing Chen
- Second Clinical College, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Jing Xiao
- Second Clinical College, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yujie Hong
- Second Clinical College, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Ke Xu
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Yan Zhu
- The Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
14
|
Wang H, Cai Y, Wu W, Zhang M, Dai Y, Wang Q. Exploring the role of gut microbiome in autoimmune diseases: A comprehensive review. Autoimmun Rev 2024; 23:103654. [PMID: 39384149 DOI: 10.1016/j.autrev.2024.103654] [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: 07/07/2024] [Revised: 09/04/2024] [Accepted: 09/28/2024] [Indexed: 10/11/2024]
Abstract
As the industrialized society advances, there has been a gradual increase in the prevalence of autoimmune disorders. A probe into the fundamental causes has disclosed several factors in modern society that have an influence on the gut microbiome. These dramatic shifts in the gut microbiome are likely to be one of the reasons for the disarray in the immune system, and the relationship between the immune system and the gut microbiome emerging as a perennial hot topic of research. This review enumerates the findings from sequencing studies of gut microbiota on seven autoimmune diseases (ADs): Rheumatoid Arthritis (RA), Systemic Lupus Erythematosus (SLE), Ankylosing Spondylitis (AS), Systemic Sclerosis (SSc), Sjögren's Syndrome (SjS), Juvenile Idiopathic Arthritis (JIA), and Behçet's Disease (BD). It aims to identify commonalities in changes in the gut microbiome within the autoimmune disease cohort and characteristics specific to each disease. The dysregulation of the gut microbiome involves a disruption of the internal balance and the balance between the external environment and the host. This dysregulation impacts the host's immune system, potentially playing a role in the development of ADs. Damage to the gut epithelial barrier allows potential pathogens to translocate to the mucosal layer, contacting epithelial cells, disrupting tight junctions, and being recognized by antigen-presenting cells, which triggers an immune response. Primed T-cells assist B-cells in producing antibodies against pathogens; if antigen mimicry occurs, an immune response is generated in extraintestinal organs during immune cell circulation, clinically manifesting as ADs. However, current research is limited; advancements in sequencing technology, large-scale cohort studies, and fecal microbiota transplantation (FMT) research are expected to propel this field to new peaks.
Collapse
Affiliation(s)
- Hongli Wang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China; The Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China
| | - Yueshu Cai
- Department of Urology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Wenqi Wu
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China; The Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China
| | - Miaomiao Zhang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China; The Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China
| | - Yong Dai
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China; The Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China
| | - Qingwen Wang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China; The Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China.
| |
Collapse
|
15
|
Zhao K, Zhao Y, Guo A, Xiao S, Tu C. Oral Microbiota Variations in Psoriasis Patients Without Comorbidity. Clin Cosmet Investig Dermatol 2024; 17:2231-2241. [PMID: 39399065 PMCID: PMC11468564 DOI: 10.2147/ccid.s473237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 08/23/2024] [Indexed: 10/15/2024]
Abstract
Background Psoriasis is a chronic inflammatory skin disease, and its etiology is still unclear. There is increasing evidence suggesting that microorganisms may trigger psoriasis. However, the relationship between psoriasis and oral microbiota remains poorly understood. Our aim is to identify differences in the composition and diversity of the oral microbiota between patients with psoriasis and healthy controls, and to discover oral microbial markers for assessing the severity of psoriasis. Methods This study recruited 20 psoriasis patients and 20 healthy individuals, collecting their saliva to analyze the composition of the oral microbiota in psoriasis patients. We employed 16S rRNA sequencing technology and utilized various methods for oral microbiome analysis, including the Shannon Index, Gini-Simpson Index, Principal Coordinates Analysis (PCoA), non-metric multidimensional scaling (NMDS), Linear discriminant analysis Effect Size (LEfSe), Wilcoxon test, and Spearman's rank correlation. Results The results showed that the alpha diversity of oral microbiota was higher in psoriasis patients. The relative abundances of certain bacterial taxa differed between psoriasis and healthy individuals, including Prevotella, Prevotella 7 and Porphyromonas gingivalis, which are increased in psoriasis. We also found a positive correlation between Alloprevotella, Porphyromonas, and Neisseria with the severity of psoriasis, while Veillonella showed a negative correlation. Conclusion In summary, this study found significant changes in the composition of the oral microbiota in patients with psoriasis. Some oral bacteria are associated with psoriasis severity. It provides a new perspective on the relationship between the oral microbiota and psoriasis.
Collapse
Affiliation(s)
- Kaidi Zhao
- Department of Dermatology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
| | - Yang Zhao
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
| | - Ao Guo
- Department of Dermatology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
| | - Shengxiang Xiao
- Department of Dermatology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
| | - Chen Tu
- Department of Dermatology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
| |
Collapse
|
16
|
Malan-Müller S, Vidal R, O'Shea E, Montero E, Figuero E, Zorrilla I, de Diego-Adeliño J, Cano M, García-Portilla MP, González-Pinto A, Leza JC. Probing the oral-brain connection: oral microbiome patterns in a large community cohort with anxiety, depression, and trauma symptoms, and periodontal outcomes. Transl Psychiatry 2024; 14:419. [PMID: 39368974 PMCID: PMC11455920 DOI: 10.1038/s41398-024-03122-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/07/2024] Open
Abstract
The role of the oral microbiome in mental health has recently been appreciated within the proposed oral-brain axis. This study examined the structure and composition of the salivary microbiome in a large-scale population-based cohort of individuals reporting mental health symptoms (n = 306) compared to mentally healthy controls (n = 164) using 16S rRNA sequencing. Mental health symptoms were evaluated using validated questionnaires and included depression, anxiety, and posttraumatic stress disorder (PTSD), with accompanying periodontal outcomes. Participants also indicated current or previous diagnoses of anxiety, depression, periodontitis, and gingivitis. Mental and periodontal health variables influenced the overall composition of the oral microbiome. PTSD symptoms correlated with a lower clr-transformed relative abundance of Haemophilus sputorum and a higher clr-transformed relative abundance of Prevotella histicola. The clr-transformed relative abundance of P. histicola was also positively associated with depressive scores and negatively associated with psychological quality of life. Anxiety disorder diagnosis was associated with a lower clr-transformed relative abundance of Neisseria elongate and a higher clr-transformed relative abundance of Oribacterium asaccharolyticum. A higher clr-transformed relative abundance of Shuttleworthia and lower clr-transformed relative abundance of Capnocytophaga were evident in those who reported a clinical periodontitis diagnosis. Higher Eggerthia and lower Haemophilus parainfluenzae clr-transformed relative abundances were associated with reported clinical periodontitis diagnoses and psychotherapeutic efficacy. Functional prediction analysis revealed a potential role for tryptophan metabolism/degradation in the oral-brain axis, which was confirmed by lower plasma serotonin levels across symptomatic groups. This study sheds light on the intricate interplay between oral microbiota, periodontal and mental health outcomes, and a potential role for tryptophan metabolism in the proposed oral-brain axis, emphasizing the need for further exploration to pave the way for novel therapeutic interventions and predicting therapeutic response.
Collapse
Affiliation(s)
- Stefanie Malan-Müller
- Department of Pharmacology and Toxicology, Faculty of Medicine, University Complutense Madrid (UCM), Madrid, Spain.
- Biomedical Research Network Centre in Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), Madrid, Spain.
- Hospital 12 de Octubre Research Institute (Imas12), Madrid, Spain.
- Instituto Universitario de Investigación Neuroquímica (IUIN-UCM), Madrid, Spain.
| | - Rebeca Vidal
- Department of Pharmacology and Toxicology, Faculty of Medicine, University Complutense Madrid (UCM), Madrid, Spain
- Hospital 12 de Octubre Research Institute (Imas12), Madrid, Spain
- Instituto Universitario de Investigación Neuroquímica (IUIN-UCM), Madrid, Spain
- Red de Investigación en Atención Primaria de Adicciones (RIAPAd-ISCIII), Madrid, Spain
| | - Esther O'Shea
- Department of Pharmacology and Toxicology, Faculty of Medicine, University Complutense Madrid (UCM), Madrid, Spain
- Hospital 12 de Octubre Research Institute (Imas12), Madrid, Spain
- Instituto Universitario de Investigación Neuroquímica (IUIN-UCM), Madrid, Spain
- Red de Investigación en Atención Primaria de Adicciones (RIAPAd-ISCIII), Madrid, Spain
| | - Eduardo Montero
- ETEP (Etiology and Therapy of Periodontal and Peri-implant Diseases) Research Group, UCM, Madrid, Spain
- Department of Dental Clinical Specialties, Faculty of Dentistry, UCM, Madrid, Spain
| | - Elena Figuero
- ETEP (Etiology and Therapy of Periodontal and Peri-implant Diseases) Research Group, UCM, Madrid, Spain
- Department of Dental Clinical Specialties, Faculty of Dentistry, UCM, Madrid, Spain
| | - Iñaki Zorrilla
- Biomedical Research Network Centre in Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), Madrid, Spain
- BIOARABA, Department of Psychiatry, Hospital Universitario de Alava, UPV/EHU, Vitoria, Spain
| | - Javier de Diego-Adeliño
- Biomedical Research Network Centre in Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), Madrid, Spain
- Sant Pau Mental Health Research Group, Institut de Recerca Sant Pau (IR Sant Pau), Barcelona, Spain
- Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Autonomous University of Barcelona (UAB), Barcelona, Spain
| | - Marta Cano
- Biomedical Research Network Centre in Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), Madrid, Spain
- Sant Pau Mental Health Research Group, Institut de Recerca Sant Pau (IR Sant Pau), Barcelona, Spain
| | - Maria Paz García-Portilla
- Biomedical Research Network Centre in Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), Madrid, Spain
- Department of Psychiatry, Universidad de Oviedo, Servicio de Psiquiatría, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Ana González-Pinto
- Biomedical Research Network Centre in Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), Madrid, Spain
- BIOARABA, Department of Psychiatry, Hospital Universitario de Alava, UPV/EHU, Vitoria, Spain
| | - Juan C Leza
- Department of Pharmacology and Toxicology, Faculty of Medicine, University Complutense Madrid (UCM), Madrid, Spain
- Biomedical Research Network Centre in Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), Madrid, Spain
- Hospital 12 de Octubre Research Institute (Imas12), Madrid, Spain
- Instituto Universitario de Investigación Neuroquímica (IUIN-UCM), Madrid, Spain
| |
Collapse
|
17
|
Ruscitti P, Nunziato M, Caso F, Scarpa R, Di Maggio F, Giacomelli R, Salvatore F. Prevention of rheumatoid arthritis using a familial predictive medicine approach. Autoimmun Rev 2024; 23:103653. [PMID: 39370029 DOI: 10.1016/j.autrev.2024.103653] [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: 05/30/2024] [Revised: 09/03/2024] [Accepted: 09/28/2024] [Indexed: 10/08/2024]
Abstract
Most of the chronic-degenerative diseases deserve a very early recognition of symptoms and signs for the earliest secondary prevention, which could be also very useful in many cases for the most precocious clinical approach. The periodic monitoring of a subject at risk of a specific disease, because of genomic predisposition by predictive medicine approach, may help to earlier detection of onset and/or the progression of the pathology itself, through intra-individual monitoring. This is particularly the case of rheumatoid arthritis (RA) for which an early diagnosis is undoubtedly the first step to ensure the most proper therapy for the patient. Thus, the earlier identification of individuals at high risk of RA could lead to ultra-preventive strategies to start for the best lifestyle performances and/or for any other effective therapeutic interventions to contrast the onset, and/or the evolution of the putative RA. This will also optimize both costs and medical resources, according to the health care policies of many countries.
Collapse
Affiliation(s)
- Piero Ruscitti
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Marcella Nunziato
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Via Sergio Pansini, 5, 80131 Napoli, NA, Italy; CEINGE - Biotecnologie Avanzate - Franco Salvatore, Via Gaetano Salvatore, 486, 80145 Napoli, Italy
| | - Francesco Caso
- Rheumatology Research Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, via S. Pansini 5, 80131 Naples, Italy
| | - Raffaele Scarpa
- Rheumatology Research Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, via S. Pansini 5, 80131 Naples, Italy
| | - Federica Di Maggio
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Via Sergio Pansini, 5, 80131 Napoli, NA, Italy; CEINGE - Biotecnologie Avanzate - Franco Salvatore, Via Gaetano Salvatore, 486, 80145 Napoli, Italy
| | - Roberto Giacomelli
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Campus Bio-Medico, Via Álvaro del Portillo 200, 00128 Rome, Italy; Rheumatology, Immunology and Clinical Medicine Unit, Department of Medicine, University of Rome "Campus Biomedico" School of Medicine, Rome, Italy.
| | - Francesco Salvatore
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Via Sergio Pansini, 5, 80131 Napoli, NA, Italy; CEINGE - Biotecnologie Avanzate - Franco Salvatore, Via Gaetano Salvatore, 486, 80145 Napoli, Italy.
| |
Collapse
|
18
|
Xu Q, Fang J, Wang Y, Lang D, Xu B. The causal relationship between gut microbiota and lower extremity deep vein thrombosis combined with pulmonary embolism. Front Microbiol 2024; 15:1301737. [PMID: 39417077 PMCID: PMC11480004 DOI: 10.3389/fmicb.2024.1301737] [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: 12/07/2023] [Accepted: 09/17/2024] [Indexed: 10/19/2024] Open
Abstract
Background Over the years, numerous studies have explored the relationship between gut microbiota and lower extremity deep vein thrombosis (LEDVT) and pulmonary embolism (PE). The present study utilized Mendelian randomization (MR) to assess the causal link between gut microbiota and LEDVT combined with PE. Methods Human gut microbiota genome-wide association study (GWAS) summary data from the MiBioGen consortium (n = 18,340) were utilized. Summary-level data on LEDVT (2,116 cases and 359,078 controls) and LEDVT combined with PE (4,319 cases and 356,875 controls) were obtained from the IEU Open GWAS project. MR analysis was conducted using the inverse variance weighted (IVW) method as the primary analysis. Additionally, MR-Egger, weighted median, weighted mode, and simple mode were employed as supplementary methods. Sensitivity analyses, including tests for heterogeneity and horizontal pleiotropy, were performed. Lastly, reverse MR analysis was performed. Results The IVW analyses revealed seven causal relationships between genetic liability in the gut microbiota and LEDVT and five causal relationships between genetic liability in the gut microbiota and LEDVT combined with PE. The intersection of these outcomes identified that the genus Butyricicoccus reduced the risk of both LEDVT and LEDVT combined with PE, while the genus Clostridium innocuum increased the risk for both conditions. Conclusion This study demonstrates that the gut microbiota is causally associated with LEDVT and LEDVT combined with PE. Our findings provide valuable insights into the underlying mechanisms and suggest potential avenues for further clinical investigations of these conditions.
Collapse
Affiliation(s)
- Qiyang Xu
- Department of General Surgery, Ningbo No.2 Hospital, Ningbo, China
| | - Jihong Fang
- Department of Emergency, Ningbo Medical Center Lihuili Hospital, The Lihuili Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yi Wang
- Department of Radiotherapy and Chemotherapy, Ningbo No.2 Hospital, Ningbo, China
| | - Dehai Lang
- Department of General Surgery, Ningbo No.2 Hospital, Ningbo, China
| | - Bin Xu
- Department of General Surgery, Ningbo No.2 Hospital, Ningbo, China
| |
Collapse
|
19
|
da Costa ALA, Soares MA, Lourenço TGB, Guimarães-Pinto K, Filardy AD, de Oliveira AM, de Luca BG, Magliano DAC, Araujo OMO, Moura L, Lopes RT, Palhares de Miranda AL, Tributino JLM, Vieira Colombo AP. Periodontal pathogen Aggregatibacter actinomycetemcomitans JP2 correlates with colonic leukocytes decrease and gut microbiome imbalance in mice. J Periodontal Res 2024; 59:961-973. [PMID: 38757372 DOI: 10.1111/jre.13288] [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: 01/18/2024] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 05/18/2024]
Abstract
AIM Evidence suggests that translocation of oral pathogens through the oral-gut axis may induce intestinal dysbiosis. This study aimed to evaluate the impact of a highly leukotoxic Aggregatibacter actinomycetemcomitans (Aa) strain on the gut microbiota, intestinal mucosal integrity and immune system in healthy mice. METHODS Eight-week-old male C57BL6 mice were divided into control (n = 16) and JP2 groups (n = 19), which received intragastric gavage with PBS and with a suspension of Aa JP2 (HK921), respectively, twice a week for 4 weeks. Colonic lamina propria, fecal material, serum, gingival tissues, and mandibles were obtained for analyses of leukocyte populations, inflammatory mediators, mucosal integrity, alveolar bone loss, and gut microbiota. Differences between groups for these parameters were examined by non-parametric tests. RESULTS The gut microbial richness and the number of colonic macrophages, neutrophils, and monocytes were significantly lower in Aa JP2-infected mice than in controls (p < .05). In contrast, infected animals showed higher abundance of Clostridiaceae, Lactobacillus taiwanensis, Helicobacter rodentium, higher levels of IL-6 expression in colonic tissues, and higher splenic MPO activity than controls (p < .05). No differences in tight junction expression, serum endotoxin levels, and colonic inflammatory cytokines were observed between groups. Infected animals presented also slightly more alveolar bone loss and gingival IL-6 levels than controls (p < .05). CONCLUSION Based on this model, intragastric administration of Aa JP2 is associated with changes in the gut ecosystem of healthy hosts, characterized by less live/recruited myeloid cells, enrichment of the gut microbiota with pathobionts and decrease in commensals. Negligible levels of colonic pro-inflammatory cytokines, and no signs of mucosal barrier disruption were related to these changes.
Collapse
Affiliation(s)
- André L A da Costa
- Oral Microbiology Laboratory, Institute of Microbiology Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Cellular Immunology Laboratory, Institute of Microbiology Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mariana A Soares
- Department of Pharmaceutical Biotechnology, Laboratory of Studies in Experimental Pharmacology, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Talita G B Lourenço
- Oral Microbiology Laboratory, Institute of Microbiology Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Kamila Guimarães-Pinto
- Cellular Immunology Laboratory, Institute of Microbiology Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alessandra D Filardy
- Cellular Immunology Laboratory, Institute of Microbiology Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Adriana Miranda de Oliveira
- Oral Microbiology Laboratory, Institute of Microbiology Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Olga M O Araujo
- Laboratory of Nuclear Instrumentation, Nuclear Engineering Program, Institute Alberto Luiz de Coimbra of Graduate and Research in Engineering, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Larissa Moura
- Laboratory of Nuclear Instrumentation, Nuclear Engineering Program, Institute Alberto Luiz de Coimbra of Graduate and Research in Engineering, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ricardo Tadeu Lopes
- Laboratory of Nuclear Instrumentation, Nuclear Engineering Program, Institute Alberto Luiz de Coimbra of Graduate and Research in Engineering, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Luisa Palhares de Miranda
- Cellular Immunology Laboratory, Institute of Microbiology Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jorge L M Tributino
- Molecular Pharmacology Laboratory, Institute of Biomedical Sciences, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Paula Vieira Colombo
- Oral Microbiology Laboratory, Institute of Microbiology Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
20
|
Holers VM, Demoruelle KM, Buckner JH, James EA, Firestein GS, Robinson WH, Steere AC, Zhang F, Norris JM, Kuhn KA, Deane KD. Distinct mucosal endotypes as initiators and drivers of rheumatoid arthritis. Nat Rev Rheumatol 2024; 20:601-613. [PMID: 39251771 DOI: 10.1038/s41584-024-01154-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2024] [Indexed: 09/11/2024]
Abstract
Rheumatoid arthritis (RA) is a potentially devastating autoimmune disease. The great majority of patients with RA are seropositive for anti-citrullinated protein antibodies (ACPAs), rheumatoid factors, or other autoantibodies. The onset of clinically apparent inflammatory arthritis meeting classification criteria (clinical RA) is preceded by ACPA seropositivity for an average of 3-5 years, a period that is designated as 'at-risk' of RA for ACPA-positive individuals who do not display signs of arthritis, or 'pre-RA' for individuals who are known to have progressed to developing clinical RA. Prior studies of individuals at-risk of RA have associated pulmonary mucosal inflammation with local production of ACPAs and rheumatoid factors, leading to development of the 'mucosal origins hypothesis'. Recent work now suggests the presence of multiple distinct mucosal site-specific mechanisms that drive RA evolution. Indicatively, subsets of individuals at-risk of RA and patients with RA harbour a faecal bacterial strain that has exhibited arthritogenic activity in animal models and that favours T helper 17 (TH17) cell responses in patients. Periodontal inflammation and oral microbiota have also been suggested to promote the development of arthritis through breaches in the mucosal barrier. Herein, we argue that mucosal sites and their associated microbial strains can contribute to RA evolution via distinct pathogenic mechanisms, which can be considered causal mucosal endotypes. Future therapies instituted for prevention in the at-risk period, or, perhaps, during clinical RA as therapeutics for active arthritis, will possibly have to address these individual mechanisms as part of precision medicine approaches.
Collapse
Affiliation(s)
- V Michael Holers
- Division of Rheumatology, University of Colorado Denver, Aurora, CO, USA.
| | | | | | | | - Gary S Firestein
- Division of Rheumatology, Allergy and Immunology, University of California San Diego, La Jolla, CA, USA
| | - William H Robinson
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, USA
- VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Allen C Steere
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Fan Zhang
- Division of Rheumatology, University of Colorado Denver, Aurora, CO, USA
| | - Jill M Norris
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
| | - Kristine A Kuhn
- Division of Rheumatology, University of Colorado Denver, Aurora, CO, USA
| | - Kevin D Deane
- Division of Rheumatology, University of Colorado Denver, Aurora, CO, USA
| |
Collapse
|
21
|
Yadav S, Sapra L, Srivastava RK. Polysaccharides to postbiotics: Nurturing bone health via modulating "gut-immune axis". Int J Biol Macromol 2024; 278:134655. [PMID: 39128750 DOI: 10.1016/j.ijbiomac.2024.134655] [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: 01/11/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
The increasing prevalence of individuals affected by bone pathologies globally has sparked catastrophic concerns. Ankylosing spondylitis, osteoporosis, rheumatoid arthritis, osteoarthritis, and fractures alone impact an estimated 1.71 billion people worldwide. The gut microbiota plays a crucial role in interacting with the host through the synthesis of a diverse range of metabolites called gut-associated metabolites (GAMs), which originate from external dietary substrates or endogenous host compounds. Many metabolic disorders have been linked to alterations in the gut microbiota's activity and composition. The development of metabolic illnesses has been linked to certain microbiota-derived metabolites, such as branched-chain amino acids, bile acids, short-chain fatty acids, tryptophan, trimethylamine N-oxide, and indole derivatives. Moreover, the modulation of gut microbiota through biotics (prebiotics, probiotics and postbiotics) presents a promising avenue for therapeutic intervention. Biotics selectively promote the growth of beneficial gut bacteria, thereby enhancing the production of GAMs with potential beneficial effects on bone metabolism. Understanding the intricate interplay between GAMs, and bone-associated genes through molecular informatics holds significant promise for early diagnosis, prognosis, and novel treatment strategies for various bone disorders.
Collapse
Affiliation(s)
- Sumedha Yadav
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Leena Sapra
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rupesh K Srivastava
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India.
| |
Collapse
|
22
|
Nichols RG, Rimal B, Hao F, Peters JM, Davenport ER, Patterson AD. Chlorpyrifos modulates the mouse gut microbiota and metabolic activity. ENVIRONMENT INTERNATIONAL 2024; 192:109022. [PMID: 39312839 DOI: 10.1016/j.envint.2024.109022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/25/2024]
Abstract
The organophosphate chlorpyrifos is a commonly used pesticide for fruits and vegetables despite its association with neurotoxicity in humans. While some studies suggest that organophosphates may impact the gut microbiota, no studies to date have investigated the direct effect of chlorpyrifos on the gut microbiota with doses that approximate environmentally relevant dietary concentrations (EPA chronic reference dose: 0.3 µg/kg/day in humans and EPA acute reference dose: 5 µg/kg/day in humans). Thus, we examined the influence of chlorpyrifos on the gut microbiota by assessment of bacterial physiology and metabolism using flow cytometry, 1H NMR-based metabolomics, and changes in the cecal microbiota community with 16S rRNA amplicon sequencing and analysis. Chlorpyrifos did not directly damage bacteria but rather perturbed bacterial metabolism. Chlorpyrifos exposure to bacteria increased the concentration of amino acids, carbohydrates, and nucleic acids. The relative abundances of Lactobacillus, Allobaculum, Roseburia, and Butyricicoccus increased after exposure to chlorpyrifos. Analyses of the 16S rRNA gene amplicon data predicted decreased amino acid biosynthesis and nucleic acid degradation and increased glycolysis which was supported by 1H NMR-based metabolomics. Collectively, these results demonstrate that environmentally relevant doses of chlorpyrifos can impact the metabolic activity of isolated gut microbes which may result in an imbalance in overall gut metabolic activity.
Collapse
Affiliation(s)
- Robert G Nichols
- Department of Biology, The Pennsylvania State University, University Park, PA, USA; One Health Microbiome Center, Huck Life Sciences Institute, University Park, PA 16802, USA
| | - Bipin Rimal
- One Health Microbiome Center, Huck Life Sciences Institute, University Park, PA 16802, USA; Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, USA
| | - Fuhua Hao
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, USA
| | - Jeffrey M Peters
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, USA
| | - Emily R Davenport
- Department of Biology, The Pennsylvania State University, University Park, PA, USA; One Health Microbiome Center, Huck Life Sciences Institute, University Park, PA 16802, USA
| | - Andrew D Patterson
- One Health Microbiome Center, Huck Life Sciences Institute, University Park, PA 16802, USA; Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
23
|
Qi P, Chen X, Tian J, Zhong K, Qi Z, Li M, Xie X. The gut homeostasis-immune system axis: novel insights into rheumatoid arthritis pathogenesis and treatment. Front Immunol 2024; 15:1482214. [PMID: 39391302 PMCID: PMC11464316 DOI: 10.3389/fimmu.2024.1482214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
Rheumatoid arthritis is a widely prevalent autoimmune bone disease that imposes a significant burden on global healthcare systems due to its increasing incidence. In recent years, attention has focused on the interaction between gut homeostasis and the immune system, particularly in relation to bone health. Dysbiosis, which refers to an imbalance in the composition and function of the gut microbiota, has been shown to drive immune dysregulation through mechanisms such as the release of pro-inflammatory metabolites, increased gut permeability, and impaired regulatory T cell function. These factors collectively contribute to immune system imbalance, promoting the onset and progression of Rheumatoid arthritis. Dysbiosis induces both local and systemic inflammatory responses, activating key pro-inflammatory cytokines such as tumor necrosis factor-alpha, Interleukin-6, and Interleukin-17, which exacerbate joint inflammation and damage. Investigating the complex interactions between gut homeostasis and immune regulation in the context of Rheumatoid arthritis pathogenesis holds promise for identifying new therapeutic targets, revealing novel mechanisms of disease progression, and offering innovative strategies for clinical treatment.
Collapse
Affiliation(s)
- Peng Qi
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xin Chen
- Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Jiexiang Tian
- Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Kexin Zhong
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Zhonghua Qi
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Menghan Li
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xingwen Xie
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou, China
| |
Collapse
|
24
|
Bakinowska E, Stańska W, Kiełbowski K, Szwedkowicz A, Boboryko D, Pawlik A. Gut Dysbiosis and Dietary Interventions in Rheumatoid Arthritis-A Narrative Review. Nutrients 2024; 16:3215. [PMID: 39339815 PMCID: PMC11435214 DOI: 10.3390/nu16183215] [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: 08/11/2024] [Revised: 09/20/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic and progressive autoimmune disease. The pathogenesis of RA is complex and involves interactions between articular cells, such as fibroblast-like synoviocytes, and immune cells. These cells secrete pro-inflammatory cytokines, chemokines, metalloproteinases and other molecules that together participate in joint degradation. The current evidence suggests the important immunoregulatory role of the gut microbiome, which can affect susceptibility to diseases and infections. An altered microbiome, a phenomenon known as gut dysbiosis, is associated with the development of inflammatory diseases. Importantly, the profile of the gut microbiome depends on dietary habits. Therefore, dietary elements and interventions can indirectly impact the progression of diseases. This review summarises the evidence on the involvement of gut dysbiosis and diet in the pathogenesis of RA.
Collapse
Affiliation(s)
- Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Wiktoria Stańska
- Department of Medical Biology, Medical University of Warsaw, 00-575 Warsaw, Poland
| | - Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Agata Szwedkowicz
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Dominika Boboryko
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| |
Collapse
|
25
|
Zhou H, Balint D, Shi Q, Vartanian T, Kriegel MA, Brito I. Lupus and inflammatory bowel disease share a common set of microbiome features distinct from other autoimmune disorders. Ann Rheum Dis 2024:ard-2024-225829. [PMID: 39299726 DOI: 10.1136/ard-2024-225829] [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: 03/20/2024] [Accepted: 09/03/2024] [Indexed: 09/22/2024]
Abstract
OBJECTIVES This study aims to elucidate the microbial signatures associated with autoimmune diseases, particularly systemic lupus erythematosus (SLE) and inflammatory bowel disease (IBD), compared with colorectal cancer (CRC), to identify unique biomarkers and shared microbial mechanisms that could inform specific treatment protocols. METHODS We analysed metagenomic datasets from patient cohorts with six autoimmune conditions-SLE, IBD, multiple sclerosis, myasthenia gravis, Graves' disease and ankylosing spondylitis-contrasting these with CRC metagenomes to delineate disease-specific microbial profiles. The study focused on identifying predictive biomarkers from species profiles and functional genes, integrating protein-protein interaction analyses to explore effector-like proteins and their targets in key signalling pathways. RESULTS Distinct microbial signatures were identified across autoimmune disorders, with notable overlaps between SLE and IBD, suggesting shared microbial underpinnings. Significant predictive biomarkers highlighted the diverse microbial influences across these conditions. Protein-protein interaction analyses revealed interactions targeting glucocorticoid signalling, antigen presentation and interleukin-12 signalling pathways, offering insights into possible common disease mechanisms. Experimental validation confirmed interactions between the host protein glucocorticoid receptor (NR3C1) and specific gut bacteria-derived proteins, which may have therapeutic implications for inflammatory disorders like SLE and IBD. CONCLUSIONS Our findings underscore the gut microbiome's critical role in autoimmune diseases, offering insights into shared and distinct microbial signatures. The study highlights the potential importance of microbial biomarkers in understanding disease mechanisms and guiding treatment strategies, paving the way for novel therapeutic approaches based on microbial profiles. TRIAL REGISTRATION NUMBER NCT02394964.
Collapse
Affiliation(s)
- Hao Zhou
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Diana Balint
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Qiaojuan Shi
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | | | - Martin A Kriegel
- Department of Translational Rheumatology and Immunology, Institute of Musculoskeletal Medicine, Münster, Germany
- Section of Rheumatology and Clinical Immunology, University Hospital Münster, Münster, Germany
- Cells in Motion Interfaculty Centre, University of Münster, Münster, Germany
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Ilana Brito
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| |
Collapse
|
26
|
Liu Y, Zhang L, Yang C, Zhi L, Steven Xu X, Yuan M. Oral microbiome diversity shapes the association between sleep duration and depression. Front Neurol 2024; 15:1442557. [PMID: 39346766 PMCID: PMC11427320 DOI: 10.3389/fneur.2024.1442557] [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: 06/08/2024] [Accepted: 08/23/2024] [Indexed: 10/01/2024] Open
Abstract
Background Emerging research suggests the relationship between the oral microbiome and sleep duration with depression, however, the precise mechanisms by which oral microbial diversity influences the sleep-depression nexus remain to be elucidated. Methods We analyzed data from 4,692 participants in the National Health and Nutrition Examination Survey (NHANES), incorporating key demographic variables, oral microbiome diversity metrics, sleep duration, and depression assessment variables. Classical multidimensional scaling facilitated dimensionality reduction, while unsupervised clustering divided participants into groups based on β-diversity dissimilarity matrices. We examined the moderating effects of oral microbiome diversity on the sleep-depression relationship by incorporating interaction terms sleep-oral microbiome diversity into multiple linear regression models. Results Our analysis revealed a U-shaped relationship between sleep duration and depression. Specifically, α-diversity was a significant moderator, with reduced diversity linked to an increased depression risk in participants with insufficient sleep. Regarding β-diversity, using both Bray-Curtis and UniFrac distance measures, Cluster 2 exhibited the strongest associations in sleep-deprived individuals (Bray-Curtis: β = 1.02, p < 0.001; Weighted UniFrac: β = 0.91, p < 0.001). In contrast, Cluster 1 displayed notable effects in individuals with excessive sleep (Bray-Curtis: β = 0.63, p = 0.008). Additionally, Cluster 3 was prominently associated with depression in sleep-deprived participants using unweighted UniFrac distance (β = 0.93, p < 0.001), and Cluster 2 was significant among those with excessive sleep across both unweighted (β = 0.80, p = 0.0004) and weighted UniFrac distances (β = 0.60, p = 0.001). Conclusion This study highlights the crucial role of oral microbiome diversity in moderating the U-shaped relationship between sleep duration and depression risk.
Collapse
Affiliation(s)
- Yan Liu
- Department of Health Data Science, Anhui Medical University, Hefei, Anhui, China
| | - Ling Zhang
- Department of Health Data Science, Anhui Medical University, Hefei, Anhui, China
| | - Can Yang
- Department of Health Data Science, Anhui Medical University, Hefei, Anhui, China
| | - Liping Zhi
- Department of Health Data Science, Anhui Medical University, Hefei, Anhui, China
| | - Xu Steven Xu
- Clinical Pharmacology and Quantitative Science, Genmab Inc., Princeton, NJ, United States
| | - Min Yuan
- Department of Health Data Science, Anhui Medical University, Hefei, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, Hefei, Anhui, China
| |
Collapse
|
27
|
Wu H, Wang L, Qiu C. Causal relationship, shared genes between rheumatoid arthritis and pulp and periapical disease: evidence from GWAS and transcriptome data. Front Immunol 2024; 15:1440753. [PMID: 39346909 PMCID: PMC11427265 DOI: 10.3389/fimmu.2024.1440753] [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: 05/30/2024] [Accepted: 08/27/2024] [Indexed: 10/01/2024] Open
Abstract
Objective Patients with rheumatoid arthritis (RA) have an increased risk of developing pulp and periapical disease (PAP), but the causal relationship and shared genetic factors between these conditions have not been explored. This study aimed to investigate the bidirectional causal relationship between RA and PAP and to analyze shared genes and pathogenic pathways. Methods We utilized GWAS data from the IEU Open GWAS Project and employed five Mendelian randomization methods (MR Egger, weighted median, inverse variance weighted, simple mode, and weighted mode) to investigate the bidirectional causal relationship between RA and PAP. Transcriptome data for RA and irreversible pulpitis (IRP) were obtained from the GEO database. Hub genes were identified through differential analysis, CytoHubba, machine learning (ML), and other methods. The immune infiltration of both diseases was analyzed using the ssGSEA method. Finally, we constructed a regulatory network for miRNAs, transcription factors, chemicals, diseases, and RNA-binding proteins based on the identified hub genes. Results RA was significantly associated with an increased risk of PAP (OR = 1.1284, 95% CI 1.0674-1.1929, p < 0.001). However, there was insufficient evidence to support the hypothesis that PAP increased the risk of RA. Integrating datasets and differential analysis identified 84 shared genes primarily involved in immune and inflammatory pathways, including the IL-17 signaling pathway, Th17 cell differentiation, and TNF signaling pathway. Using CytoHubba and three ML methods, we identified three hub genes (HLA-DRA, ITGAX, and PTPRC) that are significantly correlated and valuable for diagnosing RA and IRP. We then constructed a comprehensive regulatory network using the miRDB, miRWalk, ChipBase, hTFtarget, CTD, MalaCards, DisGeNET, and ENCORI databases. Conclusion RA may increase the risk of PAP. The three key genes, HLA-DRA, ITGAX, and PTPRC, have significant diagnostic value for both RA and IRP.
Collapse
Affiliation(s)
- Huili Wu
- Department of Endodontics, Changzhou Stomatological Hospital,
Changzhou, China
| | - Lijuan Wang
- Department of Endodontics, Changzhou Stomatological Hospital,
Changzhou, China
| | - Chenjie Qiu
- Department of General Surgery, Changzhou Hospital of Traditional Chinese
Medicine, Changzhou, China
| |
Collapse
|
28
|
Zhang X, Su R, Wang H, Wu R, Fan Y, Bin Z, Gao C, Wang C. The promise of Synovial Joint-on-a-Chip in rheumatoid arthritis. Front Immunol 2024; 15:1408501. [PMID: 39324139 PMCID: PMC11422143 DOI: 10.3389/fimmu.2024.1408501] [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: 03/28/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
Rheumatoid arthritis (RA) affects millions of people worldwide, but there are limited drugs available to treat it, so acquiring a more comprehensive comprehension of the underlying reasons and mechanisms behind inflammation is crucial, as well as developing novel therapeutic approaches to manage it and mitigate or forestall associated harm. It is evident that current in vitro models cannot faithfully replicate all aspects of joint diseases, which makes them ineffective as tools for disease research and drug testing. Organ-on-a-chip (OoC) technology is an innovative platform that can mimic the microenvironment and physiological state of living tissues more realistically than traditional methods by simulating the spatial arrangement of cells and interorgan communication. This technology allows for the precise control of fluid flow, nutrient exchange, and the transmission of physicochemical signals, such as bioelectrical, mechanical stimulation and shear force. In addition, the integration of cutting-edge technologies like sensors, 3D printing, and artificial intelligence enhances the capabilities of these models. Here, we delve into OoC models with a particular focus on Synovial Joints-on-a-Chip, where we outline their structure and function, highlighting the potential of the model to advance our understanding of RA. We integrate the actual evidence regarding various OoC models and their possible integration for multisystem disease study in RA research for the first time and introduce the prospects and opportunities of the chip in RA etiology and pathological mechanism research, drug research, disease prevention and human precision medicine. Although many challenges remain, OoC holds great promise as an in vitro model that approaches physiology and dynamics.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory for Immunomicroecology, Taiyuan, Shanxi, China
- Shanxi Province Engineering Research Center of Precision Medicine for Rheumatology, Taiyuan, Shanxi, China
| | - Rui Su
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory for Immunomicroecology, Taiyuan, Shanxi, China
- Shanxi Province Engineering Research Center of Precision Medicine for Rheumatology, Taiyuan, Shanxi, China
| | - Hui Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory for Immunomicroecology, Taiyuan, Shanxi, China
- Shanxi Province Engineering Research Center of Precision Medicine for Rheumatology, Taiyuan, Shanxi, China
| | - Ruihe Wu
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory for Immunomicroecology, Taiyuan, Shanxi, China
- Shanxi Province Engineering Research Center of Precision Medicine for Rheumatology, Taiyuan, Shanxi, China
| | - Yuxin Fan
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory for Immunomicroecology, Taiyuan, Shanxi, China
- Shanxi Province Engineering Research Center of Precision Medicine for Rheumatology, Taiyuan, Shanxi, China
| | - Zexuan Bin
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory for Immunomicroecology, Taiyuan, Shanxi, China
- Shanxi Province Engineering Research Center of Precision Medicine for Rheumatology, Taiyuan, Shanxi, China
| | - Chong Gao
- Pathology, Joint Program in Transfusion Medicine, Brigham and Women’s Hospital/Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Caihong Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory for Immunomicroecology, Taiyuan, Shanxi, China
- Shanxi Province Engineering Research Center of Precision Medicine for Rheumatology, Taiyuan, Shanxi, China
| |
Collapse
|
29
|
Ravikrishnan A, Wijaya I, Png E, Chng KR, Ho EXP, Ng AHQ, Mohamed Naim AN, Gounot JS, Guan SP, Hanqing JL, Guan L, Li C, Koh JY, de Sessions PF, Koh WP, Feng L, Ng TP, Larbi A, Maier AB, Kennedy BK, Nagarajan N. Gut metagenomes of Asian octogenarians reveal metabolic potential expansion and distinct microbial species associated with aging phenotypes. Nat Commun 2024; 15:7751. [PMID: 39237540 PMCID: PMC11377447 DOI: 10.1038/s41467-024-52097-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 08/23/2024] [Indexed: 09/07/2024] Open
Abstract
While rapid demographic changes in Asia are driving the incidence of chronic aging-related diseases, the limited availability of high-quality in vivo data hampers our ability to understand complex multi-factorial contributions, including gut microbial, to healthy aging. Leveraging a well-phenotyped cohort of community-living octogenarians in Singapore, we used deep shotgun-metagenomic sequencing for high-resolution taxonomic and functional characterization of their gut microbiomes (n = 234). Joint species-level analysis with other Asian cohorts identified distinct age-associated shifts characterized by reduction in microbial richness, and specific Alistipes and Bacteroides species enrichment (e.g., Alistipes shahii and Bacteroides xylanisolvens). Functional analysis confirmed these changes correspond to metabolic potential expansion in aging towards alternate pathways synthesizing and utilizing amino-acid precursors, vis-à-vis dominant microbial guilds producing butyrate in gut from pyruvate (e.g., Faecalibacterium prausnitzii, Roseburia inulinivorans). Extending these observations to key clinical markers helped identify >10 robust microbial associations to inflammation, cardiometabolic and liver health, including potential probiotic species (e.g., Parabacteroides goldsteinii) and pathobionts (e.g., Klebsiella pneumoniae), highlighting the microbiome's role as biomarkers and potential targets for promoting healthy aging.
Collapse
Affiliation(s)
- Aarthi Ravikrishnan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
| | - Indrik Wijaya
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
| | - Eileen Png
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
| | - Kern Rei Chng
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
| | - Eliza Xin Pei Ho
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
| | - Amanda Hui Qi Ng
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
| | - Ahmad Nazri Mohamed Naim
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
| | - Jean-Sebastien Gounot
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
| | - Shou Ping Guan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Jasinda Lee Hanqing
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Lihuan Guan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Chenhao Li
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
| | - Jia Yu Koh
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
| | - Paola Florez de Sessions
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore
| | - Woon-Puay Koh
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
- Singapore Institute for Clinical Sciences (SICS), Agency for Science Technology and Research (A*STAR), 30 Medical Drive, Brenner Centre for Molecular Medicine, Singapore, 117609, Republic of Singapore
| | - Lei Feng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Tze Pin Ng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Anis Larbi
- Singapore Immunology Network (SigN), Agency for Science Technology and Research (A*STAR), 8A Biomedical Grove, Immunos, Singapore, 138648, Republic of Singapore
| | - Andrea B Maier
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
- Department of Human Movement Sciences, @AgeAmsterdam, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Brian K Kennedy
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Niranjan Nagarajan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Republic of Singapore.
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore.
| |
Collapse
|
30
|
Beas R, Altamirano-Farfan E, Izquierdo-Veraza D, Norwood DA, Riva-Moscoso A, Godoy A, Montalvan-Sanchez EE, Ramirez M, Guifarro DA, Kitchin E, Fischer M, Kurada S. Prevalence of celiac disease in systemic lupus erythematosus, sjogren syndrome and systemic sclerosis: A systematic review and meta-analysis. Dig Liver Dis 2024; 56:1475-1482. [PMID: 38584032 DOI: 10.1016/j.dld.2024.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/17/2024] [Accepted: 03/21/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND Celiac disease (CeD) is an immune-mediated disorder affecting the small bowel, associated with genetic factors and increasing global prevalence. AIM This study explores the association between CeD, Systemic Lupus Erythematosus (SLE), primary Sjogren syndrome (pSS), and Systemic Sclerosis (SSc). METHODS A systematic review and meta-analysis were conducted following PRISMA guidelines. Searches across multiple databases yielded 2728 articles, with 15 studies selected. Data extraction included study characteristics, prevalence of CeD and CeD antibodies in SLE, pSS, and SSc. Quality assessment utilized the Newcastle-Ottawa Scale. RESULTS The meta-analysis revealed a pooled prevalence of biopsy-proven CeD in SLE, pSS, and SSc of approximately 3%. Seroprevalence of any CeD antibody in SLE, pSS, and SSc ranged from 3% to 10%. Notably, pSS exhibited the highest prevalence at 5.59%. High heterogeneity was observed in seroprevalence across autoimmune conditions. Quality assessment indicated robust methodological quality in the selected studies. CONCLUSION This study highlights a significantly higher prevalence of CeD, especially pSS, compared to the general population. The findings underscore the importance of recognizing elevated CeD antibodies in patients with SLE, pSS and SSc emphasizing the need for early detection and comprehensive care for gastrointestinal symptoms in these conditions.
Collapse
Affiliation(s)
- Renato Beas
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
| | | | | | - Dalton A Norwood
- Division of Preventive Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Ambar Godoy
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Mirian Ramirez
- Ruth Lilly Medical Library, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Daniel A Guifarro
- Internal Medicine, John H. Stroger, Jr. Hospital of Cook County, Chicago, USA
| | - Emily Kitchin
- Division of Rheumatology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Monika Fischer
- Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Satya Kurada
- Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
31
|
Yang P, Ma G, Lu T, Zhou J, Fan H, Zhang X, Fan S, Xiao X. The influence of the oral microbiota in full-term pregnant women on immune regulation during pregnancy. J Reprod Immunol 2024; 165:104298. [PMID: 39002425 DOI: 10.1016/j.jri.2024.104298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/21/2024] [Accepted: 07/04/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND This study aims to conduct a preliminary exploration of the correlation between the oral microbiota of full-term pregnant women and both local placental immunity and the systemic immune system of the mother. METHODS A total of 26 pregnant women participated in this study, with samples collected from oral swabs, placental tissue, and peripheral venous blood. High-throughput sequencing was used to examine the oral microbial community. Flow cytometry was employed to assess immune cells in placental tissue and peripheral venous blood. ELISA and Luminex liquid bead chip technology were utilized to detect cytokines in both placental tissue and peripheral venous blood. RESULTS In placental tissue, The oral microbial community is primarily negatively correlated with placental CD3+CD4+CD8+T cells and positively correlated with placental IL-5. In the peripheral blood, The oral microbial community is primarily positively correlated with maternal systemic immune parameters, including CD3+CD4+ T cells and the CD4+/CD8+ ratio, as well as positively correlated with peripheral IL-18. CONCLUSIONS The oral microbiota of full-term pregnant women participates in the regulatory function of the maternal immune system. Meanwhile, the oral microbial community may also be an important factor mediating local immune regulation in the placenta.
Collapse
Affiliation(s)
- Ping Yang
- Department of Obstetrics and Gynaecology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong Province, China
| | - Guangyu Ma
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Tong Lu
- Department of Otolaryngology, Shenzhen Long Hua District Central Hospital, Shenzhen, China
| | - Juan Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Haolong Fan
- National Key Laboratory of Science and Technology on Advanced Composites in Special Environments and Center for Composite Materials and Structures, Harbin Institute of Technology, Harbin, China
| | - Xiaowei Zhang
- Department of Obstetrics and Gynaecology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong Province, China
| | - Shangrong Fan
- Department of Obstetrics and Gynaecology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong Province, China.
| | - Xiaomin Xiao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China.
| |
Collapse
|
32
|
Tian S, Ding T, Li H. Oral microbiome in human health and diseases. MLIFE 2024; 3:367-383. [PMID: 39359681 PMCID: PMC11442140 DOI: 10.1002/mlf2.12136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 04/13/2024] [Accepted: 05/05/2024] [Indexed: 10/04/2024]
Abstract
The oral cavity contains the second-largest microbiota in the human body. The cavity's anatomically and physiologically diverse niches facilitate a wide range of symbiotic bacteria living at distinct oral sites. Consequently, the oral microbiota exhibits site specificity, with diverse species, compositions, and structures influenced by specific aspects of their placement. Variations in oral microbiota structure caused by changes in these influencing factors can impact overall health and lead to the development of diseases-not only in the oral cavity but also in organs distal to the mouth-such as cancer, cardiovascular disease, and respiratory disease. Conversely, diseases can exacerbate the imbalance of the oral microbiota, creating a vicious cycle. Understanding the heterogeneity of both the oral microbiome and individual humans is important for investigating the causal links between the oral microbiome and diseases. Additionally, understanding the intricacies of the oral microbiome's composition and regulatory factors will help identify the potential causes of related diseases and develop interventions to prevent and treat illnesses in this domain. Therefore, turning to the extant research in this field, we systematically review the relationship between oral microbiome dynamics and human diseases.
Collapse
Affiliation(s)
- Siqi Tian
- Department of Immunology and Microbiology, Zhongshan School of Medicine Sun Yat-Sen University Guangzhou China
- Key Laboratory of Tropical Diseases Control (Sun Yat-Sen University) Ministry of Education Guangzhou China
| | - Tao Ding
- Department of Immunology and Microbiology, Zhongshan School of Medicine Sun Yat-Sen University Guangzhou China
- Key Laboratory of Tropical Diseases Control (Sun Yat-Sen University) Ministry of Education Guangzhou China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University) Ministry of Education, China Guangzhou China
| | - Hui Li
- Department of Immunology and Microbiology, Zhongshan School of Medicine Sun Yat-Sen University Guangzhou China
- Key Laboratory of Tropical Diseases Control (Sun Yat-Sen University) Ministry of Education Guangzhou China
| |
Collapse
|
33
|
Chen F, Zhang D, Cheng L, Zhao D, Ye H, Zheng S, Yang Q, Han B, Wang R, Li J, Chen S. Xiaowugui decoction alleviates experimental rheumatoid arthritis by suppressing Rab5a-mediated TLR4 internalization in macrophages. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155762. [PMID: 38964156 DOI: 10.1016/j.phymed.2024.155762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/09/2024] [Accepted: 05/18/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic autoimmune disorder characterized by exacerbated synovial inflammation and joint destruction. Recent studies suggest toll-like receptor 4 (TLR4) internalization facilitate inflammatory response of macrophage. The role of TLR4 internalization in the pathogenesis of RA is unknown. PURPOSE To investigate the role and mechanism of TLR4 internalization in macrophage inflammatory response of RA and explore whether TLR4 internalization mediates the anti-arthritic effect of Xiaowugui (XWG) decoction, a patented herbal formula used in China. METHODS The co-expression of TLR4 and the internalization marker, early endosome antigen 1 (EEA1), in the synovial samples of RA patients and joint tissue of collagen-induced arthritis (CIA) mice, were evaluated using immunofluorescence. The effect of Rab5a-mediated early internalization of TLR4 on the activation induced by lipopolysaccharide (LPS) in RAW264.7 cells was investigated using small interfering RNAs that act against Rab5a. CIA was induced in Rab5a-/- mice to evaluate the role of Rab5a in vivo. The disease progression and expression of Rab5a and TLR4 in the joint tissue were evaluated in CIA mice treated with XWG. Inflammatory factors production, TLR4 internalization, and activation of downstream signaling pathways were examined in RAW264.7 cells treated with XWG in vitro. RESULTS The co-expression and co-localization of TLR4 and EEA1 were elevated in the synovial samples of RA patients and joint tissue of CIA mice. Pharmaceutical inhibition of TLR4 internalization reduced macrophages inflammatory responses induced by LPS. The co-expression and co-localization of Rab5a and TLR4 were significantly increased in macrophages treated with LPS. Silencing Rab5a reduced LPS-induced TLR4 internalization, inflammatory factors production, and phosphorylation of Jun N-terminal kinases (JNK) and p65. Genetic deletion of Rab5a inhibited TLR4 internalization and the development of arthritis in vivo. The co-expression of TLR4 and Rab5a was also elevated in the synovial samples of RA patients. XWG treatment of mice with CIA alleviated arthritis and reduced the co-expression of Rab5a and TLR4 in the joint tissue. XWG treatment of macrophage inhibited LPS-induced IL-6 and TNF-α production, co-expression of Rab5a and TLR4, and phosphorylation of JNK and p65. CONCLUSIONS Our findings highlight the pathogenic role of TLR4 internalization in patients with RA and identify a novel Rab5a-dependent internalization pathway that promotes macrophage inflammatory response. XWG treatment demonstrated outstanding therapeutic effects in experimental arthritis, and targeting the Rab5a-mediated internalization of TLR4 may be the main underlying mechanism.
Collapse
Affiliation(s)
- Feilong Chen
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China; Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, PR China
| | - Dingding Zhang
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, PR China
| | - Lifang Cheng
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Di Zhao
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China; Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, PR China
| | - Haixin Ye
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, PR China
| | - Songyuan Zheng
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Qian Yang
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Bingqi Han
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, PR China
| | - Ran Wang
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Juan Li
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China; Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, PR China.
| | - Shixian Chen
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China.
| |
Collapse
|
34
|
Vivas AJ, Boumediene S, Tobón GJ. Predicting autoimmune diseases: A comprehensive review of classic biomarkers and advances in artificial intelligence. Autoimmun Rev 2024; 23:103611. [PMID: 39209014 DOI: 10.1016/j.autrev.2024.103611] [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: 01/09/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Autoimmune diseases comprise a spectrum of disorders characterized by the dysregulation of immune tolerance, resulting in tissue or organ damage and inflammation. Their prevalence has been on the rise, significantly impacting patients' quality of life and escalating healthcare costs. Consequently, the prediction of autoimmune diseases has recently garnered substantial interest among researchers. Despite their wide heterogeneity, many autoimmune diseases exhibit a consistent pattern of paraclinical findings that hold predictive value. From serum biomarkers to various machine learning approaches, the array of available methods has been continuously expanding. The emergence of artificial intelligence (AI) presents an exciting new range of possibilities, with notable advancements already underway. The ultimate objective should revolve around disease prevention across all levels. This review provides a comprehensive summary of the most recent data pertaining to the prediction of diverse autoimmune diseases and encompasses both traditional biomarkers and the latest innovations in AI.
Collapse
Affiliation(s)
| | - Synda Boumediene
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University-School of Medicine, Springfield, IL, United States of America
| | - Gabriel J Tobón
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University-School of Medicine, Springfield, IL, United States of America; Department of Internal Medicine, Division of Rheumatology, Southern Illinois University-School of Medicine, Springfield, IL, United States of America.
| |
Collapse
|
35
|
Aminu S, Ascandari A, Laamarti M, Safdi NEH, El Allali A, Daoud R. Exploring microbial worlds: a review of whole genome sequencing and its application in characterizing the microbial communities. Crit Rev Microbiol 2024; 50:805-829. [PMID: 38006569 DOI: 10.1080/1040841x.2023.2282447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 10/20/2023] [Accepted: 11/06/2023] [Indexed: 11/27/2023]
Abstract
The classical microbiology techniques have inherent limitations in unraveling the complexity of microbial communities, necessitating the pivotal role of sequencing in studying the diversity of microbial communities. Whole genome sequencing (WGS) enables researchers to uncover the metabolic capabilities of the microbial community, providing valuable insights into the microbiome. Herein, we present an overview of the rapid advancements achieved thus far in the use of WGS in microbiome research. There was an upsurge in publications, particularly in 2021 and 2022 with the United States, China, and India leading the metagenomics research landscape. The Illumina platform has emerged as the widely adopted sequencing technology, whereas a significant focus of metagenomics has been on understanding the relationship between the gut microbiome and human health where distinct bacterial species have been linked to various diseases. Additionally, studies have explored the impact of human activities on microbial communities, including the potential spread of pathogenic bacteria and antimicrobial resistance genes in different ecosystems. Furthermore, WGS is used in investigating the microbiome of various animal species and plant tissues such as the rhizosphere microbiome. Overall, this review reflects the importance of WGS in metagenomics studies and underscores its remarkable power in illuminating the variety and intricacy of the microbiome in different environments.
Collapse
Affiliation(s)
- Suleiman Aminu
- Chemical and Biochemical Sciences-Green Process Engineering, University Mohammed VI Polytechnic, Ben Guerir, Morocco
- Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria
| | - AbdulAziz Ascandari
- Chemical and Biochemical Sciences-Green Process Engineering, University Mohammed VI Polytechnic, Ben Guerir, Morocco
| | - Meriem Laamarti
- Faculty of Medical Sciences, University Mohammed VI Polytechnic, Ben Guerir, Morocco
| | - Nour El Houda Safdi
- AgroBioSciences Program, College for Sustainable Agriculture and Environmental Science, University Mohammed VI Polytechnic, Ben Guerir, Morocco
| | - Achraf El Allali
- Bioinformatics Laboratory, College of Computing, University Mohammed VI Polytechnic, Ben Guerir, Morocco
| | - Rachid Daoud
- Chemical and Biochemical Sciences-Green Process Engineering, University Mohammed VI Polytechnic, Ben Guerir, Morocco
| |
Collapse
|
36
|
Borrego A, Koury Cabrera WH, Souza AT, Eto SF, de Oliveira SL, Rodrigues J, Jensen JR. Microbiota transfer early after birth modulates genetic susceptibility to chronic arthritis in mice. Microbes Infect 2024:105411. [PMID: 39216617 DOI: 10.1016/j.micinf.2024.105411] [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: 05/09/2024] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Genetics is central to the susceptibility or resistance to autoimmunity, and mounting evidence indicates that the intestinal microbiota also plays an essential role. In murine arthritis models, short-chain fat acid supplementation reduces disease severity by modulating tryptophan-metabolizing bacteria. Common microbiota transfer methods modulate arthritis severity, however, they are not practical for chronic models such as pristane-induced arthritis (PIA). PIA-resistant (HIII) and PIA-susceptible (LIII) mice harbor diverse intestinal microbiomes, which might be implicated in their divergent susceptibility. To investigate this hypothesis, we used cross-fostering to stably transfer the microbiota. In this study, we show that extreme susceptibility to arthritis can be modulated by early microbiota transfer, with long-lasting effects. HIII and LIII pups were cross-fostered and injected with pristane after weaning. PIA severity in cross-fostered LIII mice was significantly reduced in the chronic phase. Metagenomic analyses showed that HIII and LIII microbiomes were partly shifted by cross-fostering. Microbial groups whose abundance was associated with either HIII or LIII mice presented similar composition in cross-fostered mice of the opposite strains, suggesting a role in PIA susceptibility. Identification of bacterial groups that modulate chronic arthritis will contribute novel insights on the pathogenesis of human rheumatoid arthritis and targets for replication and functional studies.
Collapse
Affiliation(s)
- Andrea Borrego
- Laboratório de Imunogenética, Instituto Butantan, São Paulo, 05503-900, Brazil
| | | | - Alanis Tiozzo Souza
- Laboratório de Imunogenética, Instituto Butantan, São Paulo, 05503-900, Brazil
| | - Silas Fernandes Eto
- Laboratório de Desenvolvimento e Inovação, Instituto Butantan, São Paulo, 05503-900, Brazil; Center of Excellence in New Target Discovery, Instituto Butantan, São Paulo, 05503-900, Brazil
| | - Silvio Luis de Oliveira
- Setor de Microbiologia e Imunologia, Instituto de Biociências, Universidade Estadual Paulista, Botucatu, São Paulo, 18618-970, Brazil
| | - Josias Rodrigues
- Lab. de Microbioma e Genômica Bacteriana (LMGB), Instituto de Biociências, Universidade Estadual Paulista, Botucatu, São Paulo, 18618-970, Brazil
| | - José Ricardo Jensen
- Laboratório de Imunogenética, Instituto Butantan, São Paulo, 05503-900, Brazil.
| |
Collapse
|
37
|
Ranjbar M, Naeini F, Rostamian A, Djafarian K, Mohammadi H. Effects of probiotics supplementation in gastrointestinal complications and quality of life of patients with systemic sclerosis: A systematic review. Heliyon 2024; 10:e36230. [PMID: 39247342 PMCID: PMC11379610 DOI: 10.1016/j.heliyon.2024.e36230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/07/2024] [Accepted: 08/12/2024] [Indexed: 09/10/2024] Open
Abstract
Background Systemic sclerosis (SSc), as an autoimmune rheumatic disease characterized by immune dysregulation and vasculopathy, affects multiple organs. Due to the high burden of its symptoms on the health care system, this study aims to investigate the effects of probiotic supplements in patients with SSc. Methods We searched electronic databases with predefined search terms in PubMed, Scopus, and ISI Web of Science up to June 2023. Randomized controlled trials that evaluated the effects of probiotic supplementation in adult patients suffering from SSc were included in the study. Results of the included studies were reported as weighted mean difference (WMD) with a 95 % confidence interval (CI). Results Four studies met the inclusion criteria and were included in the meta-analysis. There was a total of 176 SSc patients. The results show a significant effect of probiotics supplementation on gastrointestinal (GI) symptoms containing reflux (WMD: -0.36, 95 % CI: -0.51 to -0.22, p-value <0.001), gas and bloating (WMD: -0.88, 95 % CI: -1.05 to -0.7, p-value<0.001). However, the results for constipation (WMD: -0.12, 95 % CI: -0.27 to 0.04, p-value = 0.13), diarrhea (WMD: -0.14, 95 % CI: -0.31 to 0.03, p-value = 0.10), and fecal incontinence (WMD: 0.04, 95 % CI: -0.06 to 0.15, p-value = 0.43) were insignificant. Conclusion Supplementing with probiotics may alleviate a few numbers of GI complications in SSc. Nevertheless, due to the limited number of studies, more well-designed studies are needed to strengthen these results.
Collapse
Affiliation(s)
- Mahsa Ranjbar
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Science, Tehran, Iran
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Naeini
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Science, Tehran, Iran
| | | | - Kurosh Djafarian
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Science, Tehran, Iran
- Neuroscience Institute, Sports Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Mohammadi
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Science, Tehran, Iran
| |
Collapse
|
38
|
Wang S, Liu Y, Zou X, Pan M, Wan Q, Chu X. Exploring the pathogenesis of RA through the gut-articular axis-dysbiosis a potential factor. Clin Anat 2024. [PMID: 39189295 DOI: 10.1002/ca.24215] [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: 05/29/2024] [Accepted: 08/04/2024] [Indexed: 08/28/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease with a complex etiology. It has been suggested that the pathogenesis of RA begins in the mucosa and then transitions to the joints when many factors interact, including microbial dysbiosis, inflammatory responses, and immune abnormalities at the mucosal site. Data from RA animals and patients suggest there are changes in the mucosal microflora before the onset of RA, and that dysbiosis of the mucosal ecology continues to play a role in the development of arthritis. Microbial dysbiosis of the mucosa reduces the normal barrier function of the intestinal tract, promotes inflammatory reactions in the mucosal areas of the intestines, and then activates the intestinal immune cells abnormally to produce a large number of auto-reactive antibodies that exacerbate arthritis. Current findings do not clarify whether dysbiosis is only a potential trigger for the development of RA. If it is possible to intervene in such microbial changes before the onset of RA, could the clinical symptoms of arthritis be prevented or reduced? Finding new ways to regulate gut flora composition to maintain gut barrier function is an ongoing challenge for the prevention and treatment of RA.
Collapse
Affiliation(s)
- Shuai Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Yue Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Xingyu Zou
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Mengjun Pan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Qing Wan
- Tongling Institutes for Food and Drug Control, Tongling, China
| | - Xiaoqin Chu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, China
- Engineering Technology Research Center of Modern Pharmaceutical Preparation, Anhui Province, China
| |
Collapse
|
39
|
Arnaiz-Villena A, Juarez I, Vaquero-Yuste C, Lledo T, Martin-Villa JM, Suarez-Trujillo F. Complex Interactions between the Human Major Histocompatibility Complex (MHC) and Microbiota: Their Roles in Disease Pathogenesis and Immune System Regulation. Biomedicines 2024; 12:1928. [PMID: 39200390 PMCID: PMC11352054 DOI: 10.3390/biomedicines12081928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 09/02/2024] Open
Abstract
The relationship between microbiota and the immune system is complex and characterized by the ways in which microbiota directs immune function interactions, both innate and acquired and also keeps activating the immune system throughout an individual's life. In this respect, the human Major Histocompatibility Complex (MHC, referred to as HLA in humans) plays a crucial role and is also established in self-defense against microbes by presenting microbial-derived peptides to the immune cells. However, this assumption has some unclear aspects that should be investigated. For example, how is the microbiota shaped by microbe species diversity, quantity and functions of the immune system, as well as the role and molecular mechanisms of the HLA complex during this process. There are autoimmune diseases related to both HLA and specific microbiota changes or alterations, many of which are mentioned in the present review. In addition, the HLA peptide presenting function should be put in a framework together with its linkage to diseases and also with HLA compatibility necessary for transplants to be successful. These are still quite an enigmatically statistical and phenomenological approach, but no firm pathogenic mechanisms have been described; thus, HLA's real functioning is still to be fully unveiled. After many years of HLA single-genes studies, firm pathogenesis mechanisms underlying disease linkage have been discovered. Finally, microbiota has been defined as conformed by bacteria, protozoa, archaea, fungi, and viruses; notwithstanding, endogenous viral sequences integrated into the human genome and other viral particles (obelisks) recently found in the digestive mucosa should be taken into account because they may influence both the microbiome and the immune system and their interactions. In this context, we propose to integrate these microbial-genetic particle components into the microbiome concept and designate it as "microgenobiota".
Collapse
Affiliation(s)
- Antonio Arnaiz-Villena
- Department of Immunology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (I.J.); (C.V.-Y.); (T.L.); (J.M.M.-V.); (F.S.-T.)
- Instituto de Investigacion Sanitaria Gegorio Marañon, 28009 Madrid, Spain
| | - Ignacio Juarez
- Department of Immunology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (I.J.); (C.V.-Y.); (T.L.); (J.M.M.-V.); (F.S.-T.)
- Instituto de Investigacion Sanitaria Gegorio Marañon, 28009 Madrid, Spain
| | - Christian Vaquero-Yuste
- Department of Immunology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (I.J.); (C.V.-Y.); (T.L.); (J.M.M.-V.); (F.S.-T.)
- Instituto de Investigacion Sanitaria Gegorio Marañon, 28009 Madrid, Spain
| | - Tomás Lledo
- Department of Immunology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (I.J.); (C.V.-Y.); (T.L.); (J.M.M.-V.); (F.S.-T.)
- Instituto de Investigacion Sanitaria Gegorio Marañon, 28009 Madrid, Spain
| | - José Manuel Martin-Villa
- Department of Immunology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (I.J.); (C.V.-Y.); (T.L.); (J.M.M.-V.); (F.S.-T.)
- Instituto de Investigacion Sanitaria Gegorio Marañon, 28009 Madrid, Spain
| | - Fabio Suarez-Trujillo
- Department of Immunology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (I.J.); (C.V.-Y.); (T.L.); (J.M.M.-V.); (F.S.-T.)
- Instituto de Investigacion Sanitaria Gegorio Marañon, 28009 Madrid, Spain
| |
Collapse
|
40
|
Li J, Li S, Jin J, Guo R, Jin Y, Cao L, Cai X, Rao P, Zhong Y, Xiang X, Sun X, Guo J, Hu F, Ye H, Jia Y, Xiao W, An Y, Zhang X, Xia B, Yang R, Zhou Y, Wu L, Qin J, He J, Wang J, Li Z. The aberrant tonsillar microbiota modulates autoimmune responses in rheumatoid arthritis. JCI Insight 2024; 9:e175916. [PMID: 39163137 PMCID: PMC11457857 DOI: 10.1172/jci.insight.175916] [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: 09/20/2023] [Accepted: 08/01/2024] [Indexed: 08/22/2024] Open
Abstract
Palatine tonsils are the only air-contacted lymphoid organs that constantly engage in crosstalk with commensal microorganisms and serve as the first handling sites against microbial antigens. While tonsil inflammations have been implicated in various autoimmune diseases, including rheumatoid arthritis (RA), the precise role of tonsillar microbiota in autoimmune pathogenesis remains inadequately characterized. In this study, we profiled the tonsillar microbiota and identified a notable dysbiosis in patients with RA, particularly within the Streptococcus genus. Specifically, patients with RA exhibited an enrichment of pathogenic Streptococcus species, including S. pyogenes, S. dysgalactiae, and S. agalactiae. Colonization with these bacteria significantly exacerbated arthritis severity and increased autoimmune responses in collagen-induced arthritis (CIA). Furthermore, immunization with peptides derived from these pathogenic Streptococcus species directly induced experimental arthritis. Conversely, patients with RA demonstrated a marked deficiency in commensal Streptococcus members, notably S. salivarius. Treatment of CIA mice with S. salivarius attenuated the progression of arthritis and downregulated autoimmune responses. These findings highlight a pathogenic link of tonsillar microbiota with RA, shedding light on their contribution to autoimmunity.
Collapse
Affiliation(s)
- Jing Li
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Shenghui Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
- Puensum Genetech Institute, Wuhan, China
| | - Jiayang Jin
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | | | - Yuebo Jin
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Lulu Cao
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Xuanlin Cai
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Peishi Rao
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Yan Zhong
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
- Department of Rheumatology and Immunology, The People’s Hospital of Xin Jiang Uygur Autonomous Region, Urumqi, China
| | - Xiaohong Xiang
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Xiaolin Sun
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Jianping Guo
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Fanlei Hu
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Hua Ye
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Yuan Jia
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Wenjing Xiao
- Emergency Department, Peking University People’s Hospital, Beijing, China
| | - Yuan An
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Xuan Zhang
- CAS Key Laboratory for Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - BinBin Xia
- CAS Key Laboratory for Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Rentao Yang
- Promegene Translational Research Institute, Shenzhen, China
| | - Yuanjie Zhou
- Promegene Translational Research Institute, Shenzhen, China
| | - Lijun Wu
- Department of Rheumatology and Immunology, The People’s Hospital of Xin Jiang Uygur Autonomous Region, Urumqi, China
| | - Junjie Qin
- Promegene Translational Research Institute, Shenzhen, China
| | - Jing He
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Jun Wang
- CAS Key Laboratory for Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhanguo Li
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| |
Collapse
|
41
|
Hu W, Chen S, Zou X, Chen Y, Luo J, Zhong P, Ma D. Oral microbiome, periodontal disease and systemic bone-related diseases in the era of homeostatic medicine. J Adv Res 2024:S2090-1232(24)00362-X. [PMID: 39159722 DOI: 10.1016/j.jare.2024.08.019] [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: 03/18/2024] [Revised: 08/03/2024] [Accepted: 08/12/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND Homeostasis is a state of self-regulation and dynamic equilibrium, maintaining the good physiological functions of each system in living organisms. In the oral cavity, the interaction between the host and the oral microbiome forms oral microbial homeostasis. Physiological bone remodeling and renewal can occur under the maintenance of oral microbial homeostasis. The imbalance of bone homeostasis is a key mechanism leading to the occurrence of systemic bone-related diseases. Considering the importance of oral microbial homeostasis in the maintenance of bone homeostasis, it still lacks a complete understanding of the relationship between oral microbiome, periodontal disease and systemic bone-related diseases. AIM OF REVIEW This review focuses on the homeostatic changes, pathogenic routes and potential mechanisms in the oral microbiome in periodontal disease and systemic bone-related diseases such as rheumatoid arthritis, osteoarthritis, osteoporosis and osteomyelitis. Additionally, this review discusses oral microbiome-based diagnostic approaches and explores probiotics, mesenchymal stem cells, and oral microbiome transplantation as promising treatment strategies. KEY SCIENTIFIC CONCEPTS OF REVIEW This review highlights the association between oral microbial homeostasis imbalance and systemic bone-related diseases, and highlights the possibility of remodeling oral microbial homeostasis for the prevention and treatment of systemic bone-related diseases.
Collapse
Affiliation(s)
- Weiqi Hu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, No 366 Jiangnan Avenue South, Guangzhou, Guangdong Province 510280, China
| | - Shuoling Chen
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, No 366 Jiangnan Avenue South, Guangzhou, Guangdong Province 510280, China
| | - Xianghui Zou
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, No 366 Jiangnan Avenue South, Guangzhou, Guangdong Province 510280, China
| | - Yan Chen
- Department of Pediatric Dentistry, Stomatological Hospital, School of Stomatology, Southern Medical University, No 366 Jiangnan Avenue South, Guangzhou, Guangdong Province 510280, China
| | - Jiayu Luo
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, No 366 Jiangnan Avenue South, Guangzhou, Guangdong Province 510280, China
| | - Peiliang Zhong
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, No 366 Jiangnan Avenue South, Guangzhou, Guangdong Province 510280, China
| | - Dandan Ma
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, No 366 Jiangnan Avenue South, Guangzhou, Guangdong Province 510280, China.
| |
Collapse
|
42
|
Margutti P, D’Ambrosio A, Zamboni S. Microbiota-Derived Extracellular Vesicle as Emerging Actors in Host Interactions. Int J Mol Sci 2024; 25:8722. [PMID: 39201409 PMCID: PMC11354844 DOI: 10.3390/ijms25168722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 09/02/2024] Open
Abstract
The human microbiota is an intricate micro-ecosystem comprising a diverse range of dynamic microbial populations mainly consisting of bacteria, whose interactions with hosts strongly affect several physiological and pathological processes. The gut microbiota is being increasingly recognized as a critical player in maintaining homeostasis, contributing to the main functions of the intestine and distal organs such as the brain. However, gut dysbiosis, characterized by composition and function alterations of microbiota with intestinal barrier dysfunction has been linked to the development and progression of several pathologies, including intestinal inflammatory diseases, systemic autoimmune diseases, such as rheumatic arthritis, and neurodegenerative diseases, such as Alzheimer's disease. Moreover, oral microbiota research has gained significant interest in recent years due to its potential impact on overall health. Emerging evidence on the role of microbiota-host interactions in health and disease has triggered a marked interest on the functional role of bacterial extracellular vesicles (BEVs) as mediators of inter-kingdom communication. Accumulating evidence reveals that BEVs mediate host interactions by transporting and delivering into host cells effector molecules that modulate host signaling pathways and cell processes, influencing health and disease. This review discusses the critical role of BEVs from the gut, lung, skin and oral cavity in the epithelium, immune system, and CNS interactions.
Collapse
Affiliation(s)
- Paola Margutti
- Department of Neurosciences, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.D.); (S.Z.)
| | | | | |
Collapse
|
43
|
Wu H, Yuan H, Zhang J, He T, Deng Y, Chen Y, Zhang Y, Chen W, Wu C. Helicobacter pylori upregulates PAD4 expression via stabilising HIF-1α to exacerbate rheumatoid arthritis. Ann Rheum Dis 2024:ard-2023-225306. [PMID: 39107082 DOI: 10.1136/ard-2023-225306] [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: 11/21/2023] [Accepted: 07/21/2024] [Indexed: 08/09/2024]
Abstract
OBJECTIVE Helicobacter pylori infection has been reported to aggravate rheumatoid arthritis (RA), but the relevant mechanism remains unclear. This study aimed to investigate the underlying pathogenic mechanism of H. pylori infection in the progression of RA. METHODS The Disease Activity Score (DAS-28) and serum anticitrullinated protein antibody (ACPA) levels were compared between H. pylori-negative and H. pylori-positive patients with RA. MH7A cells were stimulated with polyclonal ACPA purified from the peripheral blood of patients with RA. The citrullination levels were assessed by western blot in GES-1 cells and sera. ChIP, luciferase reporter assays, mass spectrometry and ELISA were applied to explore the molecular mechanism of H. pylori infection in RA progression. RESULTS The DAS-28 and ACPA levels of patients with RA in the H. pylori-positive group were significantly higher than those in the H. pylori-negative group. Polyclonal ACPA derived from H. pylori-positive patients promoted cell proliferation and induced secretion of IL-6 and IL-8. For the first time, we found that H. pylori infection induces cellular protein citrullination by upregulating protein arginine deiminase type 4 (PAD4). Furthermore, we confirmed a direct functional binding of hypoxia-inducible factor 1α on the PADI4 gene promoter. We demonstrated that PAD4 interacts with and citrullinates keratin 1 (K1), and serum and synovial fluid levels of anti-Cit-K1 antibody were markedly increased in H. pylori-infected patients with RA. CONCLUSION Our findings reveal a novel mechanism by which H. pylori infection contributes to RA progression. Therapeutic interventions targeting H. pylori may be a viable strategy for the management of RA.
Collapse
Affiliation(s)
- Hui Wu
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Hanmei Yuan
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Jin Zhang
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Taojun He
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Yilin Deng
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Ying Chen
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Yunqi Zhang
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, Guangdong, China
| | - Weisan Chen
- Biochemistry and Genetics, La Trobe University, Melbourne, Victoria, Australia
| | - Chao Wu
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| |
Collapse
|
44
|
Joo YB, Lee J, Park YJ, Bang SY, Kim K, Lee HS. Associations of upper respiratory mucosa microbiota with Rheumatoid arthritis, autoantibodies, and disease activity. PLoS One 2024; 19:e0308010. [PMID: 39106279 PMCID: PMC11302903 DOI: 10.1371/journal.pone.0308010] [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: 04/24/2024] [Accepted: 07/16/2024] [Indexed: 08/09/2024] Open
Abstract
The lung is recognized as a site for initiating the formation of self-antigen and autoimmune responses in rheumatoid arthritis (RA). We aimed to investigate the association of upper respiratory microbiota with RA, autoantibody production, and disease activity. Forty-six patients with RA and 17 controls were examined. Nasopharyngeal swab samples were sequenced for microbiome profiling using the V3-V4 region of the 16S rRNA gene. The microbial diversity and relative abundance were compared between RA patients and controls. Correlation analyses were conducted to evaluate the relationship between microbial abundance and clinical markers such as autoantibodies and disease activity. Microbial diversity analysis revealed no major differences between RA patients and healthy controls. However, beta diversity analysis indicated a subtle distinction in microbial composition (unweighted UniFrac distance) between the two groups (P = 0.03), hinting at a minor subset of microbiota associated with disease status. Differential abundance analysis uncovered specific taxa at various taxonomic levels, including Saccharibacteria (TM7) [O-1] (PFDR = 2.53 × 10-2), TM7 [F-1] (PFDR = 5.20 × 10-3), Microbacterium (PFDR = 3.37 × 10-4), and Stenotrophomonas (PFDR = 2.57 × 10-3). The relative abundance of ten genera correlated significantly with anti-cyclic citrullinated peptide (anti-CCP) antibody levels (PFDR < 0.05) and 11 genera were significantly associated with disease activity markers, including ESR, CRP, DAS28-ESR, and DAS-CRP (PFDR < 0.05). In particular, Saccharibacteria TM7 [G-3] and Peptostreptococcaceae [XI] [G-1] were correlated with all disease activity biomarkers. Dysbiosis in the upper respiratory mucosa is associated with RA, anti-CCP antibody levels, and disease activity.
Collapse
Affiliation(s)
- Young Bin Joo
- Department of Internal Medicine, Division of Rheumatology, Hanyang University Guri Hospital, Guri, Republic of Korea
- Hanyang University Institute for Rheumatology Research, Seoul, Republic of Korea
| | - Juho Lee
- Department of Biology, Kyung Hee University, Seoul, Republic of Korea
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Republic of Korea
| | - Yune-Jung Park
- Department of Internal Medicine, Division of Rheumatology, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - So-Young Bang
- Department of Internal Medicine, Division of Rheumatology, Hanyang University Guri Hospital, Guri, Republic of Korea
- Hanyang University Institute for Rheumatology Research, Seoul, Republic of Korea
| | - Kwangwoo Kim
- Department of Biology, Kyung Hee University, Seoul, Republic of Korea
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Republic of Korea
| | - Hye-Soon Lee
- Department of Internal Medicine, Division of Rheumatology, Hanyang University Guri Hospital, Guri, Republic of Korea
- Hanyang University Institute for Rheumatology Research, Seoul, Republic of Korea
| |
Collapse
|
45
|
Marashi A, Hasany S, Moghimi S, Kiani R, Mehran Asl S, Dareghlou YA, Lorestani P, Varmazyar S, Jafari F, Ataeian S, Naghavi K, Sajjadi SM, Haratian N, Alinezhad A, Azhdarimoghaddam A, Sadat Rafiei SK, Anar MA. Targeting gut-microbiota for gastric cancer treatment: a systematic review. Front Med (Lausanne) 2024; 11:1412709. [PMID: 39170038 PMCID: PMC11337614 DOI: 10.3389/fmed.2024.1412709] [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: 04/05/2024] [Accepted: 07/17/2024] [Indexed: 08/23/2024] Open
Abstract
Background Preclinical research has identified the mechanisms via which bacteria influence cancer treatment outcomes. Clinical studies have demonstrated the potential to modify the microbiome in cancer treatment. Herein, we systematically analyze how gut microorganisms interact with chemotherapy and immune checkpoint inhibitors, specifically focusing on how gut bacteria affect the pharmacokinetics and pharmacodynamics of cancer treatment. Method This study searched Web of Science, Scopus, and PubMed until August 2023. Studies were screened by their title and abstract using the Rayyan intelligent tool for systematic reviews. Quality assessment of studies was done using the JBI critical appraisal tool. Result Alterations in the gut microbiome are associated with gastric cancer and precancerous lesions. These alterations include reduced microbial alpha diversity, increased bacterial overgrowth, and decreased richness and evenness of gastric bacteria. Helicobacter pylori infection is associated with reduced richness and evenness of gastric bacteria, while eradication only partially restores microbial diversity. The gut microbiome also affects the response to cancer treatments, with higher abundances of Lactobacillus associated with better response to anti-PD-1/PD-L1 immunotherapy and more prolonged progression-free survival. Antibiotic-induced gut microbiota dysbiosis can reduce the anti-tumor efficacy of 5-Fluorouracil treatment, while probiotics did not significantly enhance it. A probiotic combination containing Bifidobacterium infantis, Lactobacillus acidophilus, Enterococcus faecalis, and Bacillus cereus can reduce inflammation, enhance immunity, and restore a healthier gut microbial balance in gastric cancer patients after partial gastrectomy. Conclusion Probiotics and targeted interventions to modulate the gut microbiome have shown promising results in cancer prevention and treatment efficacy.Systematic review registration: https://osf.io/6vcjp.
Collapse
Affiliation(s)
- Amir Marashi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saina Hasany
- Student Research Committee, Islamic Azad University Tehran Medical Sciences, Tehran, Iran
| | - Sadra Moghimi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Kiani
- Student Research Committee, Islamic Azad University Tehran Medical Sciences, Mashhad, Iran
| | - Sina Mehran Asl
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Student Research Committee, Islamic Azad University Tehran Medical Sciences, Tehran, Iran
| | | | - Parsa Lorestani
- School of Medicine, Shahroud Azad University of Medical Sciences, Shahroud, Iran
| | - Shirin Varmazyar
- Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Alborz, Iran
| | - Fatemeh Jafari
- School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shakiba Ataeian
- School of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Kiana Naghavi
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Negar Haratian
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arman Alinezhad
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Mahsa Asadi Anar
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
46
|
Lamba A, Taneja V. Gut microbiota as a sensor of autoimmune response and treatment for rheumatoid arthritis. Immunol Rev 2024; 325:90-106. [PMID: 38867408 PMCID: PMC11338721 DOI: 10.1111/imr.13359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
Rheumatoid arthritis (RA) is considered a multifactorial condition where interaction between the genetic and environmental factors lead to immune dysregulation causing autoreactivity. While among the various genetic factors, HLA-DR4 and DQ8, have been reported to be the strongest risk factors, the role of various environmental factors has been unclear. Though events initiating autoreactivity remain unknown, a mucosal origin of RA has gained attention based on the recent observations with the gut dysbiosis in patients. However, causality of gut dysbiosis has been difficult to prove in humans. Mouse models, especially mice expressing RA-susceptible and -resistant HLA class II genes have helped unravel the complex interactions between genetic factors and gut microbiome. This review describes the interactions between HLA genes and gut dysbiosis in sex-biased preclinical autoreactivity and discusses the potential use of endogenous commensals as indicators of treatment efficacy as well as therapeutic tool to suppress pro-inflammatory response in rheumatoid arthritis.
Collapse
Affiliation(s)
| | - Veena Taneja
- Department of Immunology and Division of Rheumatology, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
47
|
Nayak RR, Orellana DA. The impact of the human gut microbiome on the treatment of autoimmune disease. Immunol Rev 2024; 325:107-130. [PMID: 38864582 PMCID: PMC11338731 DOI: 10.1111/imr.13358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2024]
Abstract
Autoimmune (or rheumatic) diseases are increasing in prevalence but selecting the best therapy for each patient proceeds in trial-and-error fashion. This strategy can lead to ineffective therapy resulting in irreversible damage and suffering; thus, there is a need to bring the promise of precision medicine to patients with autoimmune disease. While host factors partially determine the therapeutic response to immunosuppressive drugs, these are not routinely used to tailor therapy. Thus, non-host factors likely contribute. Here, we consider the impact of the human gut microbiome in the treatment of autoimmunity. We propose that the gut microbiome can be manipulated to improve therapy and to derive greater benefit from existing therapies. We focus on the mechanisms by which the human gut microbiome impacts treatment response, provide a framework to interrogate these mechanisms, review a case study of a widely-used anti-rheumatic drug, and discuss challenges with studying multiple complex systems: the microbiome, the human immune system, and autoimmune disease. We consider open questions that remain in the field and speculate on the future of drug-microbiome-autoimmune disease interactions. Finally, we present a blue-sky vision for how the microbiome can be used to bring the promise of precision medicine to patients with rheumatic disease.
Collapse
Affiliation(s)
- Renuka R Nayak
- Rheumatology Division, Department of Medicine, University of California, San Francisco, California, USA
- Veterans Affairs Medical Center, San Francisco, California, USA
| | - Diego A Orellana
- Rheumatology Division, Department of Medicine, University of California, San Francisco, California, USA
- Veterans Affairs Medical Center, San Francisco, California, USA
| |
Collapse
|
48
|
Toyoda T, Mankia K. Prevention of Rheumatoid Arthritis in At-Risk Individuals: Current Status and Future Prospects. Drugs 2024; 84:895-907. [PMID: 38954266 DOI: 10.1007/s40265-024-02061-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2024] [Indexed: 07/04/2024]
Abstract
Early intervention has been the cornerstone of improving outcomes in patients with rheumatoid arthritis. Over the past decade, the boundaries have been pushed in an attempt to achieve effective prevention strategies in those who are at high risk of developing rheumatoid arthritis. Core risk factors including the presence of serum anti-citrullinated protein antibodies, arthralgia and subclinical inflammation on imaging are highly predictive of arthritis development. The influence of air pollution, diet and the role of microbiome on disease progression are less clear. In turn, therapeutic focus has shifted to an earlier pre-arthritis phase of the disease continuum where the clinically apparent arthritis may potentially be intercepted. Seven proof-of-concept interventional trials in at-risk individuals have been conducted so far. Whether true prevention of rheumatoid arthritis is possible remains elusive. Promising signals towards permanent disease modulation and improvement in symptom burden were seen with some immunomodulatory therapies, whilst others were unsuccessful. Long-term follow-up is required to ascertain a true effect. Looking forward, a better understanding of the natural history and underlying biological mechanisms of arthritis development and more accurate, validated risk stratification is needed.
Collapse
Affiliation(s)
- Task Toyoda
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Kulveer Mankia
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK.
| |
Collapse
|
49
|
Sharma P, Brown S, Sokoya EM. Re-evaluation of dietary interventions in rheumatoid arthritis: can we improve patient conversations around food choices? Rheumatol Int 2024; 44:1409-1419. [PMID: 38376558 PMCID: PMC11222287 DOI: 10.1007/s00296-024-05541-4] [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: 12/14/2023] [Accepted: 01/09/2024] [Indexed: 02/21/2024]
Abstract
Rheumatoid arthritis (RA) is one of over 100 different types of autoimmune conditions. In RA, the cells of the immune system attack the tissue lining the joints, triggering inflammation. A large body of research suggests that the underlying trigger(s) of RA are unique to an individual. For example, increased risk of RA can be driven by smoking tobacco in one individual and mercury exposure in another. Due to the development of next-generation sequencing technology, the critical role of the microbiota in shaping RA risk has been elucidated. Therefore, it is surprising that diet, arguably the most important lever in shaping the gut microbiota, is ineffective in the treatment of RA, even in a sub-set of patients. To attempt to rationalise this apparent paradox, we conducted an umbrella review to address the question as to whether diet can affect outcomes in RA.
Collapse
Affiliation(s)
- Prakriti Sharma
- College of Medicine and Public Health, Flinders University, Flinders Health and Medical Research Institute, Adelaide, SA, Australia
| | - Shannon Brown
- Flinders University Library, Adelaide, SA, Australia
| | - Elke M Sokoya
- College of Medicine and Public Health, Flinders University, Flinders Health and Medical Research Institute, Adelaide, SA, Australia.
| |
Collapse
|
50
|
Hu F, Li X, Liu K, Li Y, Xie Y, Wei C, Liu S, Song J, Wang P, Shi L, Li C, Li J, Xu L, Xue J, Zheng X, Bai M, Fang X, Jin X, Cao L, Hao P, He J, Wang J, Zhang C, Li Z. Rheumatoid arthritis patients harbour aberrant enteric bacteriophages with autoimmunity-provoking potential: a paired sibling study. Ann Rheum Dis 2024:ard-2024-225564. [PMID: 39084885 DOI: 10.1136/ard-2024-225564] [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: 01/22/2024] [Accepted: 07/15/2024] [Indexed: 08/02/2024]
Abstract
OBJECTIVES Viruses have been considered as important participants in the development of rheumatoid arthritis (RA). However, the profile of enteric virome and its role in RA remains elusive. This study aimed to investigate the atlas and involvement of virome in RA pathogenesis. METHODS Faecal samples from 30 pairs of RA and healthy siblings that minimise genetic interferences were collected for metagenomic sequencing. The α and β diversity of the virome and the virome-bacteriome interaction were analysed. The differential bacteriophages were identified, and their correlations with clinical and immunological features of RA were analysed. The potential involvement of these differential bacteriophages in RA pathogenesis was further investigated by auxiliary metabolic gene annotation and molecular mimicry study. The responses of CD4+ T cells and B cells to the mimotopes derived from the differential bacteriophages were systemically studied. RESULTS The composition of the enteric bacteriophageome was distorted in RA. The differentially presented bacteriophages correlated with the immunological features of RA, including anti-CCP autoantibody and HLA-DR shared epitope. Intriguingly, the glycerolipid and purine metabolic genes were highly active in the bacteriophages from RA. Moreover, peptides of RA-enriched phages, in particular Prevotella phage and Oscillibacter phage could provoke the autoimmune responses in CD4+ T cells and plasma cells via molecular mimicry of the disease-associated autoantigen epitopes, especially those of Bip. CONCLUSIONS This study provides new insights into enteric bacteriophageome in RA development. In particular, the aberrant bacteriophages demonstrated autoimmunity-provoking potential that would promote the occurrence of the disease.
Collapse
Affiliation(s)
- Fanlei Hu
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xin Li
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Kai Liu
- Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, University of Chinese Academy of Sciences, shanghai, China
- Department of Clinical Laboratory, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yanpeng Li
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yang Xie
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Chaonan Wei
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Shuyan Liu
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Jing Song
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Ping Wang
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Lianjie Shi
- Department of Rheumatology and Immunology, Peking University Shougang Hospital, Beijing, China
| | - Chun Li
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Jing Li
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Liling Xu
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Jimeng Xue
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Xi Zheng
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Mingxin Bai
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Xiangyu Fang
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Xu Jin
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Lulu Cao
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Pei Hao
- Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, University of Chinese Academy of Sciences, shanghai, China
| | - Jing He
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Jun Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Science, Beijing, China
| | - Chiyu Zhang
- Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, University of Chinese Academy of Sciences, shanghai, China
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Zhanguo Li
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| |
Collapse
|