1
|
Ning P, Lin S, Shi Y, Liu T. Potential role of gut-related factors in the pathology of cartilage in osteoarthritis. Front Nutr 2025; 11:1515806. [PMID: 39845920 PMCID: PMC11753001 DOI: 10.3389/fnut.2024.1515806] [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: 10/23/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025] Open
Abstract
Osteoarthritis (OA) is a common progressive degenerative disease. Gut microbiota (GM) and their metabolites have been closely associated with the onset, progression, and pathology of OA. GM and their metabolites may influence the cartilage directly, or indirectly by affecting the gut, the immune system, and the endocrine system. They function through classical pathways in cartilage metabolism and novel pathways that have recently been discovered. Some of them have been used as targets for the prevention and treatment of OA. The current study sought to describe the major pathological signaling pathways in OA chondrocytes and the potential role of gut-related factors in these pathways.
Collapse
Affiliation(s)
- Peng Ning
- Department of Pediatric Orthopaedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shuting Lin
- Department of Pediatric Orthopaedics, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yongyan Shi
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tianjing Liu
- Department of Pediatric Orthopaedics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
2
|
Luengo‐Gil G, Conesa‐Zamora P. Potential Utility of Induced Translocation of Engineered Bacteria as a Therapeutic Agent for Mounting a Personalized Neoantigen-Based Tumor Immune Response. GLOBAL CHALLENGES (HOBOKEN, NJ) 2022; 6:2100051. [PMID: 35284089 PMCID: PMC8902290 DOI: 10.1002/gch2.202100051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 08/16/2021] [Indexed: 06/14/2023]
Abstract
Today, an unprecedented understanding of the cancer genome, along with major breakthroughs in oncoimmunotherapy, and a resurgence of nucleic acid vaccines against cancer are being achieved. However, in most cases, the immune system response is still insufficient to react against cancer, especially in those tumors showing low mutational burden. One way to counteract tumor escape can be the induction of bacterial translocation, a phenomenon associated with autoimmune diseases which consists of a leakage in the colonic mucosa barrier, causing the access of gut bacteria to sterile body compartments such as blood. Certain commensal or live-attenuated bacteria can be engineered in such a way as to contain nucleic acids coding for tumor neoantigens previously selected from individual tumor RNAseq data. Hypothetically, these modified bacteria, previously administered orally to a cancer patient, can be translocated by several compounds acting on colonic mucosa, thus releasing neoantigens in a systemic environment in the context of an acute inflammation. Several strategies for selecting neoantigens, suitable bacteria strains, genetic constructs, and translocation inducers to achieve tumor-specific activations of CD4 and CD8 T-cells are discussed in this hypothesis.
Collapse
Affiliation(s)
- Ginés Luengo‐Gil
- Clinical Analysis and Pathology DepartmentGroup of Molecular Pathology and PharmacogeneticsInstitute for Biohealth Research from Murcia (IMIB)Hospital Universitario Santa Lucíac/Mezquita snCartagena30202Spain
| | - Pablo Conesa‐Zamora
- Clinical Analysis and Pathology DepartmentGroup of Molecular Pathology and PharmacogeneticsInstitute for Biohealth Research from Murcia (IMIB)Hospital Universitario Santa Lucíac/Mezquita snCartagena30202Spain
- Pathology and Histology DepartmentFacultad de Ciencias de la SaludUCAM Universidad Católica San Antonio de MurciaCampus de los Jerónimos, s/n, GuadalupeMurcia30107Spain
| |
Collapse
|
3
|
Tu H, Xiao E, Liu O. Taking Microbiota into Consideration in Mesenchymal Stem Cell Research. J Dent Res 2022; 101:880-886. [PMID: 35196924 DOI: 10.1177/00220345221077986] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are a promising therapy in regenerative medicine, but the clinical efficacy has yet to be identified, because the functions of MSCs are modulated by many factors, including the age and health condition of donors, origin of the tissue, and several other unknown factors. Recently, it has been revealed that, besides host factors, the microbiota that inhabits the human body is a modulator of MSCs as well. Here, we highlight the role of microbiota in the alteration of MSCs functions, with a specific focus on the self-renewal ability, multiple differentiation potential, and the immunomodulation capacity of MSCs. We also review the clinical trials and model research on the synergic and antagonistic effects of microbiota in stem cell therapy. In addition, we discuss the underlying mechanisms of the interplay between microbiota and MSCs, which are elucidated using omics approaches followed by verification experiments. As oral and maxillofacial tissues are important sources of MSCs, as well as a major access to diverse microbes, further studies are needed to elucidate these interactions in the oral field to make greater advancements in regenerative medicine.
Collapse
Affiliation(s)
- H Tu
- Hunan Key Laboratory of Oral Health Research and Human 3D Printing Engineering Research Central of Oral Care and Hunan Clinical Research Center of Oral Major Diseases and Oral Health and Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha City, Hunan Province, P.R. China
| | - E Xiao
- Beijing Maybio Pharmaceutical Biotechnology Development Co., Ltd., Changsha City, Hunan Province, P.R. China
| | - O Liu
- Hunan Key Laboratory of Oral Health Research and Human 3D Printing Engineering Research Central of Oral Care and Hunan Clinical Research Center of Oral Major Diseases and Oral Health and Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha City, Hunan Province, P.R. China
| |
Collapse
|
4
|
Berthelot JM, Lioté F, Sibilia J. Tissue microbiota: a 'secondary-self', first target of autoimmunity? Joint Bone Spine 2021; 89:105337. [PMID: 34968748 DOI: 10.1016/j.jbspin.2021.105337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2021] [Indexed: 10/19/2022]
Affiliation(s)
- Jean-Marie Berthelot
- Rheumatology Department, Nantes University Hospital, Hôtel-Dieu, Place Alexis-Ricordeau, 44093, Nantes Cedex 01, France.
| | - Frédéric Lioté
- Rheumatology Department & Inserm UMR 1132 (centre Viggo Petersen), Hôpital Lariboisière, 2 rue Ambroise Paré, F-75010 Paris, France; Université de Paris, UFR de Médecine, F-75010 Paris, France
| | - Jean Sibilia
- Service de rhumatologie, Hôpitaux Universitaires de Strasbourg, F-67000 Strasbourg, France; RESO: Centre de Référence des Maladies Autoimmunes Systémiques Rares Est Sud-Ouest, France; INSERM UMR_S1109, Université de Strasbourg, F-67000 Strasbourg, France
| |
Collapse
|
5
|
Trautmann A. [Mechanisms underlying chronic fatigue, a symptom too often overlooked]. Med Sci (Paris) 2021; 37:910-919. [PMID: 34647880 DOI: 10.1051/medsci/2021143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Acute fatigue after exertion, like acute inflammation after injury, is useful for our body. On the contrary, both chronic fatigue and chronic inflammation are deleterious, and they are associated in many diseases. In this first part, we will analyze different immune phenomena (bystander activation, memory of the innate immune system, link with the intestinal microbiota) involved in triggering chronic inflammation. This review aims at looking for links between different signs and symptoms associated with chronic fatigue, as well as between different diseases in which severe chronic fatigue can manifest. Possible underlying mechanisms for these phenomena are discussed. This is a proposal made by a researcher, with no clinical experience, to doctors confronted with an entity that is still largely mysterious. The link between chronic inflammation, neuroinflammation and fatigue will be examined in a second part.
Collapse
Affiliation(s)
- Alain Trautmann
- UMR CNRS 8104, Inserm 1016, université Paris Descartes, Institut Cochin, rue Méchain, 75014 Paris, France
| |
Collapse
|
6
|
Zhong X, Zhang F, Yin X, Cao H, Wang X, Liu D, Chen J, Chen X. Bone Homeostasis and Gut Microbial-Dependent Signaling Pathways. J Microbiol Biotechnol 2021; 31:765-774. [PMID: 34176870 PMCID: PMC9705830 DOI: 10.4014/jmb.2104.04016] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/27/2021] [Accepted: 06/13/2021] [Indexed: 12/15/2022]
Abstract
Although research on the osteal signaling pathway has progressed, understanding of gut microbial-dependent signaling pathways for metabolic and immune bone homeostasis remains elusive. In recent years, the study of gut microbiota has shed light on our understanding of bone homeostasis. Here, we review microbiota-mediated gut-bone crosstalk via bone morphogenetic protein/SMADs, Wnt and OPG/receptor activator of nuclear factor-kappa B ligand signaling pathways in direct (translocation) and indirect (metabolite) manners. The mechanisms underlying gut microbiota involvement in these signaling pathways are relevant in immune responses, secretion of hormones, fate of osteoblasts and osteoclasts and absorption of calcium. Collectively, we propose a signaling network for maintaining a dynamic homeostasis between the skeletal system and the gut ecosystem. Additionally, the role of gut microbial improvement by dietary intervention in osteal signaling pathways has also been elucidated. This review provides unique resources from the gut microbial perspective for the discovery of new strategies for further improving treatment of bone diseases by increasing the abundance of targeted gut microbiota.
Collapse
Affiliation(s)
- Xiaohui Zhong
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, P.R. China
| | - Feng Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, P.R. China,Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi 214125, P.R. China,Nutritional Department, Affiliated Hospital of Jiangnan University, Wuxi 214125, P.R. China
| | - Xinyao Yin
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, P.R. China
| | - Hong Cao
- Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi 214125, P.R. China,Department of Endocrinology, Affiliated Hospital of Jiangnan University, Wuxi 214125, P.R. China,Nutritional Department, Affiliated Hospital of Jiangnan University, Wuxi 214125, P.R. China
| | - Xuesong Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, P.R. China,Department of Orthopedics, Affiliated Hospital of Jiangnan University, Wuxi 214125, P.R.China
| | - Dongsong Liu
- Department of Orthopedics, Affiliated Hospital of Jiangnan University, Wuxi 214125, P.R.China
| | - Jing Chen
- Department of Orthopedics, Affiliated Hospital of Jiangnan University, Wuxi 214125, P.R.China
| | - Xue Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, P.R. China,Corresponding author Phone: +86-15861589177 E-mail:
| |
Collapse
|
7
|
Berthelot JM, Darrieutort-Laffite C, Trang C, Maugars Y, Le Goff B. Contribution of mycobiota to the pathogenesis of spondyloarthritis. Joint Bone Spine 2021; 88:105245. [PMID: 34166798 DOI: 10.1016/j.jbspin.2021.105245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/09/2021] [Indexed: 12/18/2022]
Abstract
This review lists current evidences for a contribution of gut mycobiota to the pathogenesis of SpA and related conditions. Gut mycobiota has a small size as compared to bacterial microbiota, but an even greater inter- and intra-individual variability. Although most fungi (brought by food or air) are only transitory present, a core mycobiota of gut resident fungi exists, and interplays with bacteria in a complex manner. A dysbiosis of this gut mycobiota has been observed in Crohn's disease and sclerosing cholangitis, with decreased proportion of Saccharomyces cerevisiae and outgrowth of more pathogenic gut fungi. Fungal-induced lower number of commensal gut bacteria can promote translocation of some bacterial/fungal antigens through mucosae, and live fungi can also cross the epithelial border in Crohn's disease. This dysbiosis also lower the ability of bacteria to metabolize tryptophan into regulatory metabolites, consequently enhancing tryptophan metabolism within human cells, which might contribute to fatigue. Translocation of mycobiotal antigens like curdlan (beta-glucan), which plays a major role in the pathogenesis of SpA in the SGK mice, has been observed in humans. This translocation of fungal antigens in human SpA might account for the anti-Saccharomyces antibodies found in this setting. Contribution of fungal antigens to psoriasis and hidradenitis suppurativa would fit with the preferential homing of fungi in the skin area most involved in those conditions. Fungal antigens also possess autoimmune uveitis-promoting function. As genes associated with SpA (CARD9 and IL23R) strongly regulate the innate immune response against fungi, further studies on fungi contribution to SpA are needed.
Collapse
Affiliation(s)
- Jean-Marie Berthelot
- Service de rhumatologie, Hôtel-Dieu, CHU de Nantes, place Alexis-Ricordeau, 44093 Nantes cedex 01, France.
| | | | - Caroline Trang
- Service de gastro-entérologie, Hôtel-Dieu, CHU de Nantes, place Alexis-Ricordeau, 44093 Nantes cedex 01, France
| | - Yves Maugars
- Service de rhumatologie, Hôtel-Dieu, CHU de Nantes, place Alexis-Ricordeau, 44093 Nantes cedex 01, France
| | - Benoît Le Goff
- Service de rhumatologie, Hôtel-Dieu, CHU de Nantes, place Alexis-Ricordeau, 44093 Nantes cedex 01, France
| |
Collapse
|
8
|
Bordagaray MJ, Fernández A, Garrido M, Astorga J, Hoare A, Hernández M. Systemic and Extraradicular Bacterial Translocation in Apical Periodontitis. Front Cell Infect Microbiol 2021; 11:649925. [PMID: 33816354 PMCID: PMC8017189 DOI: 10.3389/fcimb.2021.649925] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/01/2021] [Indexed: 12/19/2022] Open
Abstract
Apical periodontitis is an inflammatory disease of microbial etiology. It has been suggested that endodontic bacterial DNA might translocate to distant organs via blood vessels, but no studies have been conducted. We aimed first to explore overall extraradicular infection, as well as specifically by Porphyromonas spp; and their potential to translocate from infected root canals to blood through peripheral blood mononuclear cells. In this cross-sectional study, healthy individuals with and without a diagnosis of apical periodontitis with an associated apical lesion of endodontic origin (both, symptomatic and asymptomatic) were included. Apical lesions (N=64) were collected from volunteers with an indication of tooth extraction. Intracanal samples (N=39) and respective peripheral blood mononuclear cells from apical periodontitis (n=14) individuals with an indication of endodontic treatment, as well as from healthy individuals (n=14) were collected. The detection frequencies and loads (DNA copies/mg or DNA copies/μL) of total bacteria, Porphyromonas endodontalis and Porphyromonas gingivalis were measured by qPCR. In apical lesions, the detection frequencies (%) and median bacterial loads (DNA copies/mg) respectively were 70.8% and 4521.6 for total bacteria; 21.5% and 1789.7 for Porphyromonas endodontalis; and 18.4% and 1493.9 for Porphyromonas gingivalis. In intracanal exudates, the detection frequencies and median bacterial loads respectively were 100% and 21089.2 (DNA copies/μL) for total bacteria, 41% and 8263.9 for Porphyromonas endodontalis; and 20.5%, median 12538.9 for Porphyromonas gingivalis. Finally, bacteria were detected in all samples of peripheral blood mononuclear cells including apical periodontitis and healthy groups, though total bacterial loads (median DNA copies/μL) were significantly higher in apical periodontitis (953.6) compared to controls (300.7), p<0.05. Porphyromonas endodontalis was equally detected in both groups (50%), but its bacterial load tended to be higher in apical periodontitis (262.3) than controls (158.8), p>0.05; Porphyromonas gingivalis was not detected. Bacteria and specifically Porphyromonas spp. were frequently detected in endodontic canals and apical lesions. Also, total bacteria and Porphyromonas endodontalis DNA were detected in peripheral blood mononuclear cells, supporting their plausible role in bacterial systemic translocation.
Collapse
Affiliation(s)
- María José Bordagaray
- Laboratory of Periodontal Biology, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.,Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Alejandra Fernández
- Laboratory of Periodontal Biology, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.,Faculty of Dentistry, Universidad Andres Bello, Santiago, Chile
| | - Mauricio Garrido
- Laboratory of Periodontal Biology, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.,Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Jessica Astorga
- Laboratory of Periodontal Biology, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Anilei Hoare
- Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.,Laboratory of Oral Microbiology, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Marcela Hernández
- Laboratory of Periodontal Biology, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.,Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| |
Collapse
|
9
|
Deng Y, Ge X, Li Y, Zou B, Wen X, Chen W, Lu L, Zhang M, Zhang X, Li C, Zhao C, Lin X, Zhang X, Huang X, Li X, Jin M, Peng GH, Wang D, Wang X, Lai W, Liang J, Li JJ, Liang Q, Yang L, Zhang Q, Li Y, Lu P, Hu X, Li X, Deng X, Liu Y, Zou Y, Guo S, Chen T, Qin Y, Yang F, Miao L, Chen W, Chan CC, Lin H, Liu Y, Lee RWJ, Wei L. Identification of an intraocular microbiota. Cell Discov 2021; 7:13. [PMID: 33750767 PMCID: PMC7943566 DOI: 10.1038/s41421-021-00245-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 01/26/2021] [Indexed: 12/27/2022] Open
Abstract
The current dogma in ophthalmology and vision research presumes the intraocular environment to be sterile. However, recent evidence of intestinal bacterial translocation into the bloodstream and many other internal organs including the eyes, found in healthy and diseased animal models, suggests that the intraocular cavity may also be inhabited by a microbial community. Here, we tested intraocular samples from over 1000 human eyes. Using quantitative PCR, negative staining transmission electron microscopy, direct culture, and high-throughput sequencing technologies, we demonstrated the presence of intraocular bacteria. The possibility that the microbiome from these low-biomass communities could be a contamination from other tissues and reagents was carefully evaluated and excluded. We also provide preliminary evidence that a disease-specific microbial signature characterized the intraocular environment of patients with age-related macular degeneration and glaucoma, suggesting that either spontaneous or pathogenic bacterial translocation may be associated with these common sight-threatening conditions. Furthermore, we revealed the presence of an intraocular microbiome in normal eyes from non-human mammals and demonstrated that this varied across species (rat, rabbit, pig, and macaque) and was established after birth. These findings represent the first-ever evidence of intraocular microbiota in humans.
Collapse
Affiliation(s)
- Yuhua Deng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Xiaofei Ge
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Yan Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Bin Zou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Xiaofeng Wen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Weirong Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Lin Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Meifen Zhang
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xiaomin Zhang
- Tianjin Medical University Eye Hospital, Eye Institute & School of Optometry and Ophthalmology, Tianjin, 300384, China
| | - Chunmei Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Chan Zhao
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xiaofeng Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Xiulan Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Xinhua Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Xiaorong Li
- Tianjin Medical University Eye Hospital, Eye Institute & School of Optometry and Ophthalmology, Tianjin, 300384, China
| | - Ming Jin
- Department of Ophthalmology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Guang-Hua Peng
- Department of Pathophysiology, Basic Medical College of Zhengzhou University, Zhengzhou, He'nan, 450001, China
- Department of Ophthalmology, General Hospital of Chinese People's Liberation Army, Beijing, 100853, China
| | - Dongni Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Xun Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Weiyi Lai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Juanran Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Jing Jing Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Qiaoxing Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Liu Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Qinfen Zhang
- State Key Laboratory of Biocontrol, MOE Key Laboratory of Aquatic Product Safety, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510275, China
| | - Yinyin Li
- State Key Laboratory of Biocontrol, MOE Key Laboratory of Aquatic Product Safety, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510275, China
| | - Ping Lu
- State Key Laboratory of Biocontrol, MOE Key Laboratory of Aquatic Product Safety, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510275, China
| | - Xiao Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Xifang Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Xiuli Deng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Yu Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Yanli Zou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Shixin Guo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Tingting Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Yali Qin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Fuhua Yang
- Tianjin Medical University Eye Hospital, Eye Institute & School of Optometry and Ophthalmology, Tianjin, 300384, China
| | - Li Miao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Wei Chen
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, 15261, USA
- Division of Pulmonary Medicine, Allergy and Immunology, Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, 15224, USA
| | - Chi-Chao Chan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Haotian Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Yizhi Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China.
| | - Richard W J Lee
- Translational Health Sciences, University of Bristol, Bristol, UK.
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK.
| | - Lai Wei
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China.
| |
Collapse
|
10
|
Revisiting the gut-joint axis: links between gut inflammation and spondyloarthritis. Nat Rev Rheumatol 2020; 16:415-433. [PMID: 32661321 DOI: 10.1038/s41584-020-0454-9] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2020] [Indexed: 02/07/2023]
Abstract
Gut inflammation is strongly associated with spondyloarthritis (SpA), as exemplified by the high prevalence of inflammatory bowel disease (IBD) and the even higher occurrence of subclinical gut inflammation in patients with SpA. The gut-joint axis of inflammation in SpA is further reinforced by similarities in immunopathogenesis at both anatomical sites and by the clinical success of therapies blocking TNF and IL-23 in IBD and in some forms of SpA. Many genetic risk factors are shared between SpA and IBD, and changes in the composition of gut microbiota are seen in both diseases. Current dogma is that inflammation in SpA initiates in the gut and leads to joint inflammation; however, although conceptually attractive, some research does not support this causal relationship. For example, therapies targeting IL-17A are efficacious in the joint but not the gut, and interfering with gut trafficking by targeting molecules such as α4β7 in IBD can lead to onset or flares of SpA. Several important knowledge gaps remain that must be addressed in future studies. Determining the true nature of the gut-joint axis has real-world implications for the treatment of patients with co-incident IBD and SpA and for the repurposing of therapeutics from one disease to the other.
Collapse
|
11
|
Cintio M, Scarsella E, Sgorlon S, Sandri M, Stefanon B. Gut Microbiome of Healthy and Arthritic Dogs. Vet Sci 2020; 7:vetsci7030092. [PMID: 32674496 PMCID: PMC7558702 DOI: 10.3390/vetsci7030092] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/20/2020] [Accepted: 07/11/2020] [Indexed: 12/13/2022] Open
Abstract
Several studies have underlined the interplay among host-microbiome and pathophysiological conditions of animals. Research has also focused specifically on whether and how changes in the gut microbiome have provoked the occurrence of pathological phenomena affecting cartilage and joints in humans and in laboratory animals. Here, we tried to evaluate the relationship between the gut microbiome and the hip and elbow arthritis in owned dogs. The study included 14 dogs suffering from chronic arthritis (AD) and 13 healthy dogs (HD). After the first visit and during the period of the study, the dogs, under the supervision of the owner, were fed a semi-moist complete diet supplemented with omega 3 fatty acids. Feces and blood samples were collected in the clinic at the first visit (T0) and after days (T45). The plasma C-reactive protein (CRP) was higher, and the serum vitamin B12 and folate concentrations were lower (p < 0.05) in the AD group in comparison to the HD group. Data of the fecal microbiome showed that the relative abundances of the genus Megamonas were higher in AD (p < 0.001), while the relative abundance of the families Paraprevotellaceae, Porphyromonadaceae, and Mogibacteriaceae was significantly lower in comparison to HD. The results of the study identified several bacterial groups that differed significantly in the fecal microbiome between healthy and diseased dogs. If the observed differences in fecal bacterial composition predispose dogs to hip and elbow arthritis or if these differences reflect a correlation with these conditions deserves further investigation.
Collapse
|
12
|
Expansion of Rare and Harmful Lineages is Associated with Established Rheumatoid Arthritis. J Clin Med 2020; 9:jcm9041044. [PMID: 32272752 PMCID: PMC7230781 DOI: 10.3390/jcm9041044] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 04/04/2020] [Accepted: 04/06/2020] [Indexed: 12/14/2022] Open
Abstract
Objectives: To characterize the gut microbiota profile in rheumatoid arthritis (RA) patients and investigate its association with certain characteristics of RA. Patients and methods: A nested case–control cohort of 40 patients with RA and 40 sex-age matched controls was studied. Subjects with diabetes, with any other inflammatory disease, practicing extreme diets, taking antibiotics, probiotics or under any new treatment for at least three months prior to sampling were excluded. The microbiota composition was determined by 16S rRNA pyrosequencing and bioinformatics analysis by Quantitative Insights Into Microbial Ecology (QIIME). Other variables included clinical-laboratory variables and average Disease Activity Score 28 points during the follow-up period. Multiple linear regression models were constructed to investigate the possible risk factors for the microbiota. Results: β-diversity data showed that patients tend to differ from healthy subjects according to their microbiota (p = 0.07). The analysis showed an increase in Collinsella aerofaciens, Sedimentibacter and Enterococcus genera in patients compared to controls, as well as a decrease in Dorea formicigenerans. Likewise, an increase in the activity of arginine deiminase was observed, which was found in approximately 90% of the RA genes of the genus Collinsela. The sequence number of Collinsella aerofaciens was independently associated with age (B (95%CI), −0.347 (−21.6, −2.1)), high ACPA (0.323 (27.4–390.0)) and smoking (0.300 (8.8–256.4)) in RA patients. In addition, we observed decreases in Sarcina, 02d06 and Porphyromonas bacterial lineages. Conclusion: Patients with RA present dysbiosis, resulting from an abundance of certain bacterial lineages and a decrease in others. These alterations could influence the maintenance of autoimmunity to this disease.
Collapse
|
13
|
Rossini M, Epis OM, Tinazzi I, Grembiale RD, Iagnocco A. Role of the IL-23 pathway in the pathogenesis and treatment of enthesitis in psoriatic arthritis. Expert Opin Biol Ther 2020; 20:787-798. [PMID: 32129102 DOI: 10.1080/14712598.2020.1737855] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Enthesitis is a key feature of spondyloarthritis (SpA). Several studies have underlined the role of interleukin (IL)-23 in SpA development as a crucial cytokine in the pathogenesis of enthesitis. AREA COVERED This review summarizes recent evidence of the role of IL-23 in the pathogenesis of and as a target of the treatment of enthesitis. We review the definition, diagnosis and clinical impact of enthesitis and its connection with microbial infections, gut dysbiosis, and mechanical stress. We also review clinical trials and real-life studies of drugs targeting the p19 or p40 subunits of IL-23. EXPERT OPINION Novel therapies targeting the p19 or p40 subunit of IL-23 appear to be promising treatment options for patients with enthesitis. Although we are currently unable to identify the best therapeutic window to target IL-23 in SpA disease evolution, the promising ability of this therapy to control the gut-entheseal axis is increasing our knowledge of SpA pathogenesis.
Collapse
Affiliation(s)
- Maurizio Rossini
- Rheumatology Section, Department of Medicine, University of Verona , Verona, Italy
| | | | - Ilaria Tinazzi
- Unit of Rheumatology, IRCCS Sacro Cuore Don Calabria Hospital , Verona, Italy
| | | | - Annamaria Iagnocco
- Academic Rheumatology Centre, Università Degli Studi Di Torino , Turin, Italy
| |
Collapse
|
14
|
Berthelot JM, Le Goff B, Maugars Y. Bone marrow mesenchymal stem cells in rheumatoid arthritis, spondyloarthritis, and ankylosing spondylitis: problems rather than solutions? Arthritis Res Ther 2019; 21:239. [PMID: 31722720 PMCID: PMC6854713 DOI: 10.1186/s13075-019-2014-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/24/2019] [Indexed: 12/14/2022] Open
Abstract
Background Bone marrow mesenchymal stem cells (BM-MSCs) can dampen inflammation in animal models of inflammatory rheumatisms and human osteoarthritis. They are expected to be a solution for numerous human conditions. However, in rheumatoid arthritis (RA) and spondyloarthritis (SpA), subsets of subchondral BM-MSCs might conversely fuel synovitis and enthesitis. Main text Abnormal behaviour of BM-MSCs and/or their progeny has been found in RA and SpA. BM-MSCs also contribute to the ossifying processes observed in ankylosing spondylitis. Some synovial fibroblastic stem cells probably derive from BM-MSCs, but some stem cells can also migrate through the bare zone area of joints, not covered by cartilage, into the synovium. BM-MSCs can also migrate in the synovium over tendons. Sub-populations of bone marrow stem cells also invade the soft tissue side of enthesis via small holes in the bone cortex. The present review aims (1) to make a focus on these two aspects and (2) to put forward the hypothesis that lasting epigenetic changes of some BM-MSCs, induced by transient infections of the bone marrow close to the synovium and/or entheses (i.e. trained immunity of BM-MSCs and/or their progeny), contribute to the pathogenesis of inflammatory rheumatisms. Such hypothesis would fit with (1) the uneven distribution and/or flares of arthritis and enthesitis observed at the individual level in RA and SpA (reminiscent of what is observed following reactive arthritis and/or in Whipple’s disease); (2) the subchondral bone marrow oedema and erosions occurring in many RA patients, in the bare zone area; and (3) the frequent relapses of RA and SpA despite bone marrow transplantation, whereas most BM-MSCs resist graft preconditioning. Conclusion Some BM-MSCs might be more the problem than the solution in inflammatory rheumatisms. Subchondral bone marrow BM-MSCs and their progeny trafficking through the bare zone area of joints or holes in the bone cortex of entheses should be thoroughly studied in RA and SpA respectively. This may be done first in animal models. Mini-arthroscopy of joints could also be used in humans to specifically sample tissues close to the bare zone and/or enthesis areas.
Collapse
Affiliation(s)
| | - Benoit Le Goff
- Centre Hospitalier Universitaire de Nantes, Nantes, France
| | - Yves Maugars
- Centre Hospitalier Universitaire de Nantes, Nantes, France
| |
Collapse
|
15
|
The Dynamic Interplay between the Gut Microbiota and Autoimmune Diseases. J Immunol Res 2019; 2019:7546047. [PMID: 31772949 PMCID: PMC6854958 DOI: 10.1155/2019/7546047] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/14/2019] [Indexed: 12/11/2022] Open
Abstract
The human gut-resident commensal microbiota is a unique ecosystem associated with various bodily functions, especially immunity. Gut microbiota dysbiosis plays a crucial role in autoimmune disease pathogenesis as well as in bowel-related diseases. However, the role of the gut microbiota, which causes or influences systemic immunity in autoimmune diseases, remains elusive. Aryl hydrocarbon receptor, a ligand-activated transcription factor, is a master moderator of host-microbiota interactions because it shapes the immune system and impacts host metabolism. In addition, treatment optimization while minimizing potential adverse effects in autoimmune diseases remains essential, and modulation of the gut microbiota constitutes a potential clinical therapy. Here, we present evidence linking gut microbiota dysbiosis with autoimmune mechanisms involved in disease development to identify future effective approaches based on the gut microbiota for preventing autoimmune diseases.
Collapse
|
16
|
Abstract
The human gut-resident commensal microbiota is a unique ecosystem associated with various bodily functions, especially immunity. Gut microbiota dysbiosis plays a crucial role in autoimmune disease pathogenesis as well as in bowel-related diseases. However, the role of the gut microbiota, which causes or influences systemic immunity in autoimmune diseases, remains elusive. Aryl hydrocarbon receptor, a ligand-activated transcription factor, is a master moderator of host-microbiota interactions because it shapes the immune system and impacts host metabolism. In addition, treatment optimization while minimizing potential adverse effects in autoimmune diseases remains essential, and modulation of the gut microbiota constitutes a potential clinical therapy. Here, we present evidence linking gut microbiota dysbiosis with autoimmune mechanisms involved in disease development to identify future effective approaches based on the gut microbiota for preventing autoimmune diseases.
Collapse
|