1
|
Fehringer M, Vogl T. Molecular mimicry in the pathogenesis of autoimmune rheumatic diseases. J Transl Autoimmun 2025; 10:100269. [PMID: 39877080 PMCID: PMC11773492 DOI: 10.1016/j.jtauto.2025.100269] [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: 10/31/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 01/31/2025] Open
Abstract
Autoimmune rheumatic diseases (ARDs) are a heterogeneous group of conditions characterized by excessive and misdirected immune responses against the body's own musculoskeletal tissues. Their exact aetiology remains unclear, with genetic, demographic, behavioural and environmental factors implicated in disease onset. One prominent hypothesis for the initial breach of immune tolerance (leading to autoimmunity) is molecular mimicry, which describes structural or sequence similarities between human and microbial proteins (mimotopes). This similarity can lead to cross-reactive antibodies and T-cell receptors, resulting in an immune response against autoantigens. Both commensal microbes in the human microbiome and pathogens can trigger molecular mimicry, thereby potentially contributing to the onset of ARDs. In this review, we focus on the role of molecular mimicry in the onset of rheumatoid arthritis and systemic lupus erythematosus. Moreover, implications of molecular mimicry are also briefly discussed for ankylosing spondylitis, systemic sclerosis and myositis.
Collapse
Affiliation(s)
| | - Thomas Vogl
- Medical University of Vienna, Borschkegasse 8a, 1090, Vienna, Austria
| |
Collapse
|
2
|
Ranganathan Y, Kumar PR, Paramasivam SG, Krishnan RS. A Review of Connecting Bioinformatic Techniques to Rheumatoid Arthritis and its Associated Comorbidities. Curr Rheumatol Rev 2025; 21:25-36. [PMID: 38803169 DOI: 10.2174/0115733971302188240515075547] [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: 01/23/2024] [Revised: 04/09/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024]
Abstract
Rheumatoid Arthritis (RA) is a progressive autoimmune condition inflicting serious threats to people's life and health by causing severe pain and joint destruction. It affects not only bones and joints but also causes comorbid conditions and shortens the lifetime. The interactions and synergistic effects of comorbid disease with RA are not yet well studied. Hence, understanding how these conditions will collectively affect the progression and outcome of RA is the current area of research. Identification of RA and comorbidities associated with target genes may uncover diagnosis and treatment methodologies. This review is to provide an overview of the interlinking approach of Rheumatoid Arthritis with its comorbid conditions and its systemic complications using bioinformatic techniques which would be useful to identify the genes and pathways that are in common for both RA and comorbid diseases. It would also emphasize the significance of bioinformatics in comparing the pathological features of RA and comorbid diseases. With the help of bioinformatics, valuable insights into the mechanism underlying Rheumatoid arthritis and comorbid diseases would be better understood.
Collapse
Affiliation(s)
- Yeswanth Ranganathan
- Department of Biotechnology, University College of Engineering, BIT Campus, Anna University, Tiruchirappalli, 620024, India
| | - Pritam Ramesh Kumar
- Department of Biotechnology, University College of Engineering, BIT Campus, Anna University, Tiruchirappalli, 620024, India
| | - Sudhakar Gandhi Paramasivam
- Department of Biotechnology, University College of Engineering, BIT Campus, Anna University, Tiruchirappalli, 620024, India
| | - Ravi Shankar Krishnan
- Department of Biotechnology, University College of Engineering, BIT Campus, Anna University, Tiruchirappalli, 620024, India
| |
Collapse
|
3
|
Chasov V, Gilyazova E, Ganeeva I, Zmievskaya E, Davletshin D, Valiullina A, Bulatov E. Gut Microbiota Modulation: A Novel Strategy for Rheumatoid Arthritis Therapy. Biomolecules 2024; 14:1653. [PMID: 39766360 PMCID: PMC11674688 DOI: 10.3390/biom14121653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 12/14/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that leads to joint inflammation, progressive tissue damage and significant disability, severely impacting patients' quality of life. While the exact mechanisms underlying RA remain elusive, growing evidence suggests a strong link between intestinal microbiota dysbiosis and the disease's development and progression. Differences in microbial composition between healthy individuals and RA patients point to the role of gut microbiota in modulating immune responses and promoting inflammation. Therapies targeting microbiota restoration have demonstrated promise in improving treatment efficacy, enhancing patient outcomes and slowing disease progression. However, the complex interplay between gut microbiota and autoimmune pathways in RA requires further investigation to establish causative relationships and mechanisms. Here, we review the current understanding of the gut microbiota's role in RA pathogenesis and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Vitaly Chasov
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
| | - Elvina Gilyazova
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
| | - Irina Ganeeva
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
| | - Ekaterina Zmievskaya
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
| | - Damir Davletshin
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
| | - Aygul Valiullina
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
| | - Emil Bulatov
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| |
Collapse
|
4
|
Bixio R, Bertelle D, Bertoldo E, Morciano A, Rossini M. The potential pathogenic role of gut microbiota in rheumatic diseases: a human-centred narrative review. Intern Emerg Med 2024; 19:891-900. [PMID: 38141117 DOI: 10.1007/s11739-023-03496-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023]
Abstract
A growing amount of evidence suggests that gut microbiota plays an important role in human health, including a possible role in the pathogenesis of rheumatic and musculoskeletal diseases (RMD). We analysed the current evidence about the role of microbiota in rheumatoid arthritis (RA), spondyloarthritis (SpA), systemic lupus erythematosus (SLE) and systemic sclerosis (SSc). In RA, we found a general consensus regarding a reduction of diversity and a specific bacterial signature, with consistent changes according to the different ethnic and geographical areas. The major pathogenetic role in RA is recognised for P. copri, L. salivarius and Collinsella, even if findings become more heterogeneous when considering established disease. In SpA, we found a relative gut abundance of Akkermansia, Coprococcus, Ruminoccocus and a relative reduction in Bacterioides and Firmicutes spp. Human and preclinical data suggest loss of mucosal barrier, increased permeability and Th1- and Th17-mediated inflammation. Additionally, HLA-B27 seems to play a role in shaping the intestinal microbiota and the consequent inflammation. In SLE, the typical gut microbiota signature was characterised by a reduction in the Firmicutes/Bacteroidetes ratio and by enrichment of Rhodococcus, Eggerthella, Klebsiella, Prevotella, Eubacterium and Flavonifractor, even if their real pathogenic impact remains unclear. In SSc, gastrointestinal dysbiosis is well documented with an increase of pro-inflammatory species (Fusobacterium, Prevotella, Ruminococcus, Akkermansia, γ-Proteobacteria, Erwinia, Trabsulsiella, Bifidobacterium, Lactobacillus, Firmicutes and Actinobacteria) and a reduction of species as Faecalibacterium, Clostridium, Bacteroidetes and Rikenella. In conclusion, seems possible to recognise a distinct gut microbiota profile for each RMD, even if significant differences in bacterial species do exist between different studies and there is a high risk of bias due to the cross-sectional nature of such studies. Therefore longitudinal studies are needed, especially on patients with preclinical and early disease, to investigate the real role of gut microbiota in the pathogenesis of RMD.
Collapse
Affiliation(s)
- Riccardo Bixio
- Rheumatology Section, Department of Medicine, University of Verona, P.Le L.A. Scuro 10, 37134, Verona, Italy.
| | - Davide Bertelle
- Rheumatology Section, Department of Medicine, University of Verona, P.Le L.A. Scuro 10, 37134, Verona, Italy
- Rheumatology Section, Department of Medicine, Azienda Ospedaliera Friuli Occidentale, Pordenone, Italy
| | - Eugenia Bertoldo
- Rheumatology Section, Department of Medicine, University of Verona, P.Le L.A. Scuro 10, 37134, Verona, Italy
- Internal Medicine Unit, Department of Medicine, Mater Salutis Hospital, Legnago, Italy
| | - Andrea Morciano
- Rheumatology Section, Department of Medicine, University of Verona, P.Le L.A. Scuro 10, 37134, Verona, Italy
| | - Maurizio Rossini
- Rheumatology Section, Department of Medicine, University of Verona, P.Le L.A. Scuro 10, 37134, Verona, Italy
| |
Collapse
|
5
|
Ria F, Delogu G, Ingrosso L, Sali M, Di Sante G. Secrets and lies of host-microbial interactions: MHC restriction and trans-regulation of T cell trafficking conceal the role of microbial agents on the edge between health and multifactorial/complex diseases. Cell Mol Life Sci 2024; 81:40. [PMID: 38216734 PMCID: PMC11071949 DOI: 10.1007/s00018-023-05040-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 10/04/2023] [Accepted: 11/06/2023] [Indexed: 01/14/2024]
Abstract
Here we critically discuss data supporting the view that microbial agents (pathogens, pathobionts or commensals alike) play a relevant role in the pathogenesis of multifactorial diseases, but their role is concealed by the rules presiding over T cell antigen recognition and trafficking. These rules make it difficult to associate univocally infectious agents to diseases' pathogenesis using the paradigm developed for canonical infectious diseases. (Cross-)recognition of a variable repertoire of epitopes leads to the possibility that distinct infectious agents can determine the same disease(s). There can be the need for sequential infection/colonization by two or more microorganisms to develop a given disease. Altered spreading of infectious agents can determine an unwanted activation of T cells towards a pro-inflammatory and trafficking phenotype, due to differences in the local microenvironment. Finally, trans-regulation of T cell trafficking allows infectious agents unrelated to the specificity of T cell to modify their homing to target organs, thereby driving flares of disease. The relevant role of microbial agents in largely prevalent diseases provides a conceptual basis for the evaluation of more specific therapeutic approaches, targeted to prevent (vaccine) or cure (antibiotics and/or Biologic Response Modifiers) multifactorial diseases.
Collapse
Affiliation(s)
- F Ria
- Department of Translational Medicine and Surgery, Section of General Pathology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - G Delogu
- Mater Olbia Hospital, 07026, Olbia, Italy
- Department of Biotechnological, Basic, Intensivological and Perioperatory Sciences-Section of Microbiology, Università Cattolica del S Cuore, 00168, Rome, Italy
| | - L Ingrosso
- Department Infectious Diseases, Istituto Superiore di Sanità, 00161, Rome, Italy
- European Program for Public Health Microbiology Training (EUPHEM), European Centre for Disease Prevention and Control (ECDC), Stockholm, Sweden
| | - M Sali
- Department of Biotechnological, Basic, Intensivological and Perioperatory Sciences-Section of Microbiology, Università Cattolica del S Cuore, 00168, Rome, Italy
- Department of Laboratory and Infectivology Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| | - G Di Sante
- Department of Medicine and Surgery, Section of Human, Clinical and Forensic Anatomy, University of Perugia, 60132, Perugia, Italy.
| |
Collapse
|
6
|
Abutayyem H, Alam MK, Kanwal B, Alswairki HJ, Alogaibi YA. Sterilizing orthodontic appliances: A systematic review and meta-analysis on the available methods. J Orthod Sci 2023; 12:51. [PMID: 37881658 PMCID: PMC10597368 DOI: 10.4103/jos.jos_53_23] [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: 04/10/2023] [Revised: 04/27/2023] [Accepted: 04/27/2023] [Indexed: 10/27/2023] Open
Abstract
Infection control is essential to protect both the doctor and the patient by preventing the spread of infectious diseases. There is no exception in the field of dentistry, particularly in orthodontics, where numerous appliances are used for a variety of functions and also because the mouth cavity has the highest concentration of bacteria of any body part. Through this systematic review, we aimed to assess the various methods of sterilization employed in an orthodontic setting. Using relevant keywords, reference searches, and citation searches, the databases such as PubMed, MEDLINE, Web of Science, Cochrane, and Scopus were all searched; a total of 206 documents were found, of which 113 were initially selected. The remaining 23 distinct papers were initially made available after 90 publications that were identical to or similar to one another were eliminated. The final selection was made from eight documents that met all inclusion and exclusion requirements. The existing methods of sterilization were found to be competent in dealing with the microorganisms found in a typical orthodontic setting. The chemical method of sterilization was the norm in most of the studies that we assessed, with glutaraldehyde and peracetic acid (PAA) being the most commonly employed compounds for disinfection. PROSPERO Registration Number: CRD42022380831.
Collapse
Affiliation(s)
- Huda Abutayyem
- Department of Clinical Sciences, Center of Medical and Bio-Allied Health Sciences Research, College of Dentistry, Ajman University, Ajman, United Arab Emirates
| | - Mohammad Khursheed Alam
- Orthodontic Division, Preventive Dentistry Department, Orthodontic Division, College of Dentistry, Jouf University, Sakaka, Saudi Arabia
- Department of Dental Research Cell, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
- Department of Public Health, Faculty of Allied Health Sciences, Daffodil lnternational University, Ashulia, Dhaka, Bangladesh
| | - Bushra Kanwal
- Orthodontic Specialist, Practicing in Dental Clinic, AlBaha, Saudi Arabia
| | | | - Yahya A. Alogaibi
- Orthodontic Consultant, Aseer Specialized Dental Center, Abha, Saudi Arabia
| |
Collapse
|
7
|
Romero-Figueroa MDS, Ramírez-Durán N, Montiel-Jarquín AJ, Horta-Baas G. Gut-joint axis: Gut dysbiosis can contribute to the onset of rheumatoid arthritis via multiple pathways. Front Cell Infect Microbiol 2023; 13:1092118. [PMID: 36779190 PMCID: PMC9911673 DOI: 10.3389/fcimb.2023.1092118] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/16/2023] [Indexed: 02/14/2023] Open
Abstract
Rheumatoid Arthritis (RA) is an autoimmune disease characterized by loss of immune tolerance and chronic inflammation. It is pathogenesis complex and includes interaction between genetic and environmental factors. Current evidence supports the hypothesis that gut dysbiosis may play the role of environmental triggers of arthritis in animals and humans. Progress in the understanding of the gut microbiome and RA. has been remarkable in the last decade. In vitro and in vivo experiments revealed that gut dysbiosis could shape the immune system and cause persistent immune inflammatory responses. Furthermore, gut dysbiosis could induce alterations in intestinal permeability, which have been found to predate arthritis onset. In contrast, metabolites derived from the intestinal microbiota have an immunomodulatory and anti-inflammatory effect. However, the precise underlying mechanisms by which gut dysbiosis induces the development of arthritis remain elusive. This review aimed to highlight the mechanisms by which gut dysbiosis could contribute to the pathogenesis of RA. The overall data showed that gut dysbiosis could contribute to RA pathogenesis by multiple pathways, including alterations in gut barrier function, molecular mimicry, gut dysbiosis influences the activation and the differentiation of innate and acquired immune cells, cross-talk between gut microbiota-derived metabolites and immune cells, and alterations in the microenvironment. The relative weight of each of these mechanisms in RA pathogenesis remains uncertain. Recent studies showed a substantial role for gut microbiota-derived metabolites pathway, especially butyrate, in the RA pathogenesis.
Collapse
Affiliation(s)
| | - Ninfa Ramírez-Durán
- Laboratory of Medical and Environmental Microbiology, Department of Medicine, Autonomous University of the State of Mexico, Toluca, Mexico
| | - Alvaro José Montiel-Jarquín
- Dirección de Educación e Investigación en Salud, Hospital de Especialidades de Puebla, Instituto Mexicano del Seguro Social, Puebla, Mexico
| | - Gabriel Horta-Baas
- Rheumatology Service, Internal Medicine Department, Instituto Mexicano del Seguro Social, Merida, Mexico
| |
Collapse
|
8
|
A Comprehensive Review of the Current and Future Role of the Microbiome in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2022; 14:cancers14041020. [PMID: 35205769 PMCID: PMC8870349 DOI: 10.3390/cancers14041020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/11/2022] [Accepted: 02/15/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary This review summarizes the current literature related to the microbiome and pancreatic ductal adenocarcinoma (PDAC). The aim of this review is to explore the current role of the microbiome in the disease process, screening/diagnostics and to postulate the future role with regards to therapeutic strategies including chemotherapy, immunotherapy and surgery. We further explore the future of microbiome modulation (faecal microbiome transplants, bacterial consortiums, anti-microbials and probiotics), their applications and how we can improve the future of microbiome modulation in a bid to improve PDAC outcomes. Abstract Pancreatic ductal adenocarcinoma (PDAC) is expected to become the second most common cause of cancer death in the USA by 2030, yet progress continues to lag behind that of other cancers, with only 9% of patients surviving beyond 5 years. Long-term survivorship of PDAC and improving survival has, until recently, escaped our understanding. One recent frontier in the cancer field is the microbiome. The microbiome collectively refers to the extensive community of bacteria and fungi that colonise us. It is estimated that there is one to ten prokaryotic cells for each human somatic cell, yet, the significance of this community in health and disease has, until recently, been overlooked. This review examines the role of the microbiome in PDAC and how it may alter survival outcomes. We evaluate the possibility of employing microbiomic signatures as biomarkers of PDAC. Ultimately this review analyses whether the microbiome may be amenable to targeting and consequently altering the natural history of PDAC.
Collapse
|
9
|
Di Sante G, Gremese E, Tolusso B, Cattani P, Di Mario C, Marchetti S, Alivernini S, Tredicine M, Petricca L, Palucci I, Camponeschi C, Aragon V, Gambotto A, Ria F, Ferraccioli G. Haemophilus parasuis ( Glaesserella parasuis) as a Potential Driver of Molecular Mimicry and Inflammation in Rheumatoid Arthritis. Front Med (Lausanne) 2021; 8:671018. [PMID: 34485325 PMCID: PMC8415917 DOI: 10.3389/fmed.2021.671018] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 07/19/2021] [Indexed: 01/07/2023] Open
Abstract
Background:Haemophilus parasuis (Hps; now Glaesserella parasuis) is an infectious agent that causes severe arthritis in swines and shares sequence similarity with residues 261–273 of collagen type 2 (Coll261−273), a possible autoantigen in rheumatoid arthritis (RA). Objectives/methods: We tested the presence of Hps sequencing 16S ribosomal RNA in crevicular fluid, synovial fluids, and tissues in patients with arthritis (RA and other peripheral arthritides) and in healthy controls. Moreover, we examined the cross-recognition of Hps by Coll261−273-specific T cells in HLA-DRB1*04pos RA patients, by T-cell receptor (TCR) beta chain spectratyping and T-cell phenotyping. Results:Hps DNA was present in 57.4% of the tooth crevicular fluids of RA patients and in 31.6% of controls. Anti-Hps IgM and IgG titers were detectable and correlated with disease duration and the age of the patients. Peripheral blood mononuclear cells (PBMCs) were stimulated with Hps virulence-associated trimeric autotransporter peptide (VtaA10755−766), homologous to human Coll261−273 or co-cultured with live Hps. In both conditions, the expanded TCR repertoire overlapped with Coll261−273 and led to the production of IL-17. Discussion: We show that the DNA of an infectious agent (Hps), not previously described as pathogen in humans, is present in most patients with RA and that an Hps peptide is able to activate T cells specific for Coll261−273, likely inducing or maintaining a molecular mimicry mechanism. Conclusion: The cross-reactivity between VtaA10755−766 of a non-human infectious agent and human Coll261−273 suggests an involvement in the pathogenesis of RA. This mechanism appears emphasized in predisposed individuals, such as patients with shared epitope.
Collapse
Affiliation(s)
- Gabriele Di Sante
- Section of General Pathology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Elisa Gremese
- Division of Rheumatology, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Rome, Italy.,Division of Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Barbara Tolusso
- Division of Rheumatology, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Rome, Italy
| | - Paola Cattani
- Dipartimento di Scienze di laboratorio e infettivologiche, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy.,Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Sezione di Microbiologia, Università Cattolica del S. Cuore, Rome, Italy
| | - Clara Di Mario
- Division of Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Simona Marchetti
- Dipartimento di Scienze di laboratorio e infettivologiche, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Stefano Alivernini
- Division of Rheumatology, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Rome, Italy
| | - Maria Tredicine
- Section of General Pathology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Luca Petricca
- Division of Rheumatology, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Rome, Italy
| | - Ivana Palucci
- Dipartimento di Scienze di laboratorio e infettivologiche, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy.,Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Sezione di Microbiologia, Università Cattolica del S. Cuore, Rome, Italy
| | - Chiara Camponeschi
- Section of General Pathology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Virginia Aragon
- Institut de Recerca i Tecnologies Agroalimentaries, Centre de Recerca en Sanitat Animal (CReSA IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Andrea Gambotto
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Department of Molecular Genetics and Biochemistry, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Francesco Ria
- Section of General Pathology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy.,Dipartimento di Scienze di laboratorio e infettivologiche, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | | |
Collapse
|
10
|
Genetic Factors of Predisposition and Clinical Characteristics of Rheumatoid Arthritis in Russian Patients. J Pers Med 2021; 11:jpm11060469. [PMID: 34070522 PMCID: PMC8228085 DOI: 10.3390/jpm11060469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/21/2021] [Accepted: 05/21/2021] [Indexed: 12/14/2022] Open
Abstract
Rheumatoid arthritis (RA) is a multifactorial disease caused by a genetic predisposition and environmental factors. Predisposing alleles of various genes have a relatively small influence on the disease risk when they appear separately, but in combination, they predispose an individual to RA development. We genotyped 125 patients with RA including 60 SNPs and sequenced coding part of six genes by next-generation sequencing (NGS) technology on a target panel (IAD177464_185). According to our data, the alleles HLA-DRB1*04, HLA-DRB1*01, HLA-B*27, PTPN22 (rs2476601), TNF (rs1800629), TPMT (rs2842934), and IL4 (rs2243250), and genotypes HLA-DRB1*04:04, HLA-DRB1*01:16, PTPN22 (rs2476601), TPMT (rs2842934), were significantly associated with the RA development. Associations with clinical criteria (DAS28-CRP, HAQ-DI, and CDAI) and biochemical factors were investigated. We have shown that the PADI4 genotypes (rs11203367, rs2240340, rs11203366, and rs874881) are significantly associated with the baseline levels of DAS28-CRP, HAQ-DI, and CDAI; genotypes IL23R (rs7530511) and TNFRSF1A (rs748004, rs2228144) with the level of anti citrullinated peptide antibodies (ACPA); the genotypes DHODH (rs3213422) and MTHFR (rs180113) with the concentration of C-reactive protein (CRP); and the genotypes IL2RA (rs2104286), IRAK3 (rs11541076), and IL4R (rs1801275) with the level of rheumatoid factor (RF). Application of targeted NGS panel contributes to expanded genotyping to identify risk groups among the RA patients.
Collapse
|
11
|
Wu X, Chen X, Lyu X, Zheng H. Advances in Microbiome Detection Technologies and Application in Antirheumatic Drug Design. Curr Pharm Des 2021; 27:891-899. [PMID: 33308114 DOI: 10.2174/1381612826666201211114609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/25/2020] [Indexed: 11/22/2022]
Abstract
Rheumatic diseases are a kind of chronic inflammatory and autoimmune disease affecting the connection or supporting structures of the human body, such as the most common diseases Ankylosing spondylitis (AS), gout and Systemic lupus erythematosus (SLE). Although the precise etiology and pathogenesis of the different types of rheumatic diseases remain mostly unknown, it is now commonly believed that these diseases are attributed to some complex interactions between genetics and environmental factors, especially the gut microbiome. Altered microbiome showed clinical improvement in disease symptoms and partially restored to normality after prescribing disease-modifying antirheumatic drugs (DMARDs) or other treatment strategies. Recent advances in next-generation sequencing-based microbial profiling technology, especially metagenomics, have identified alteration of the composition and function of the gut microbiota in patients. Clinical and experimental data suggest that dysbiosis may play a pivotal role in the pathogenesis of these diseases. In this paper, we provide a brief review of the advances in the microbial profiling technology and up-to-date resources for accurate taxonomic assignment of metagenomic reads, which is a key step for metagenomics studies. In addition, we review the altered gut microbiota signatures that have been reported so far across various studies, upon which diagnostics classification models can be constructed, and the drug-induced regulation of the host microbiota can be used to control disease progression and symptoms.
Collapse
Affiliation(s)
- Xin Wu
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, China
| | - Xiang Chen
- Department of Bioinformatics, Hangzhou Nuowei Information Technology, Co., Ltd. Hangzhou, China
| | - Xiaochen Lyu
- Department of Bioinformatics, Hangzhou Nuowei Information Technology, Co., Ltd. Hangzhou, China
| | - Hao Zheng
- Department of Bioinformatics, Hangzhou Nuowei Information Technology, Co., Ltd. Hangzhou, China
| |
Collapse
|
12
|
Mangalam AK, Yadav M, Yadav R. The Emerging World of Microbiome in Autoimmune Disorders: Opportunities and Challenges. INDIAN JOURNAL OF RHEUMATOLOGY 2021; 16:57-72. [PMID: 34531642 PMCID: PMC8442979 DOI: 10.4103/injr.injr_210_20] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Trillions of commensal bacteria colonizing humans (microbiome) have emerged as essential player(s) in human health. The alteration of the same has been linked with diseases including autoimmune disorders such as multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, and ankylosing spondylitis. Gut bacteria are separated from the host through a physical barrier such as skin or gut epithelial lining. However, the perturbation in the healthy bacterial community (gut dysbiosis) can compromise gut barrier integrity, resulting in translocation of bacterial contents across the epithelial barrier (leaky gut). Bacterial contents such as lipopolysaccharide and bacterial antigens can induce a systemic inflammatory environment through activation and induction of immune cells. The biggest question in the field is whether inflammation causes gut dysbiosis or dysbiosis leads to disease induction or propagation, i.e., it is inside out or outside in or both. In this review, we first discuss the microbiome profiling studies in various autoimmune disorders, followed by a discussion of potential mechanisms through which microbiome is involved in the pathobiology of diseases. A better understanding of the role of the microbiome in health and disease will help us harness the power of commensal bacteria for the development of novel therapeutic agents to treat autoimmune disorders.
Collapse
Affiliation(s)
| | - Meeta Yadav
- Department of Pathology, University of Iowa, Iowa, IA,
USA
| | - Rajwardhan Yadav
- Department of Rheumatology, St Francis Hospital, Hartford,
CT, USA
| |
Collapse
|
13
|
Durlik-Popińska K, Żarnowiec P, Lechowicz Ł, Gawęda J, Kaca W. Antibodies Isolated from Rheumatoid Arthritis Patients against Lysine-Containing Proteus mirabilis O3 (S1959) Lipopolysaccharide May React with Collagen Type I. Int J Mol Sci 2020; 21:ijms21249635. [PMID: 33348817 PMCID: PMC7767033 DOI: 10.3390/ijms21249635] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/10/2020] [Accepted: 12/14/2020] [Indexed: 02/03/2023] Open
Abstract
Most rheumatic diseases, including rheumatoid arthritis (RA), are characterized by immune disorders that affect antibody activity. In the present study, using Dot blot and ELISA assay, we showed that patients with rheumatic disease produced significantly more antibodies against lipopolysaccharide (LPS) P. mirabilis O3 compared to healthy donors (p < 0.05), and affinity purified antibodies against LPS O3 may cross-react with collagen type I. It was demonstrated that purified of antibodies isolated from RA patients sera, reacted stronger with the collagen than healthy donors (p = 0.015), and cross-reaction was correlated with level of anti-citrullinated peptide antibodies (r = 0.7, p = 0.003). Moreover, using six different lipopolysaccharides were demonstrated the significant correlations in sera reactivity among lysine-containing lipopolysaccharides observed in patients’ sera (p < 0.05). Using Attenuated Total Reflection Fourier Transform Infrared Spectroscopy (ATR-FTIR) it was shown that unique wavenumbers of sera spectra correlate with reactivity with lipopolysaccharides allowing distinguish patients from healthy blood donors. Antibodies adsorption by synthetic antigens shows that in patients’ group anti-LPS O3 antibodies can be adsorbed by both amides of galacturonic acid and lysine or threonine, which suggests less specificity of antibodies binding with non-carbohydrate LPS component. The observed correlations suggest that non-carbohydrate components of LPS may be an important epitope for less specific anti-LPS antibodies, which might lead to cross-reactions and affect disease development.
Collapse
Affiliation(s)
- Katarzyna Durlik-Popińska
- Department of Microbiology and Parasitology, Institute of Biology, Jan Kochanowski University, 25-369 Kielce, Poland; (P.Ż.); (Ł.L.); (W.K.)
- Correspondence:
| | - Paulina Żarnowiec
- Department of Microbiology and Parasitology, Institute of Biology, Jan Kochanowski University, 25-369 Kielce, Poland; (P.Ż.); (Ł.L.); (W.K.)
| | - Łukasz Lechowicz
- Department of Microbiology and Parasitology, Institute of Biology, Jan Kochanowski University, 25-369 Kielce, Poland; (P.Ż.); (Ł.L.); (W.K.)
| | - Józef Gawęda
- Rheumatology Clinic ARTIMED, 25-022 Kielce, Poland;
| | - Wiesław Kaca
- Department of Microbiology and Parasitology, Institute of Biology, Jan Kochanowski University, 25-369 Kielce, Poland; (P.Ż.); (Ł.L.); (W.K.)
| |
Collapse
|
14
|
Huang R, Li F, Zhou Y, Zeng Z, He X, Fang L, Pan F, Chen Y, Lin J, Li J, Qiu D, Tian Y, Tan X, Song Y, Xu Y, Lai Y, Yi H, Gao Q, Fang X, Shi M, Zhou C, Huang J, He YT. Metagenome-wide association study of the alterations in the intestinal microbiome composition of ankylosing spondylitis patients and the effect of traditional and herbal treatment. J Med Microbiol 2020; 69:797-805. [PMID: 31778109 PMCID: PMC7451032 DOI: 10.1099/jmm.0.001107] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 10/23/2019] [Indexed: 01/07/2023] Open
Abstract
Introduction. Ankylosing spondylitis (AS) is a systemic progressive disease with an unknown etiology that may be related to the gut microbiome. Therefore, a more thorough understanding of its pathogenesis is necessary for directing future therapy.Aim. We aimed to determine the differences in intestinal microbial composition between healthy individuals and patients with AS who received and who did not receive treatment interventions. In parallel, the pathology of AS in each patient was analysed to better understand the link between AS treatment and the intestinal microbiota of the patients.Methodology. Sixty-six faecal DNA samples, including 37 from healthy controls (HCs), 11 from patients with untreated AS (NM), 7 from patients treated with nonsteroidal anti-inflammatory drugs (e.g. celecoxib; WM) and 11 from patients treated with Chinese herbal medicine (CHM), such as the Bushen-Qiangdu-Zhilv decoction, were collected and used in the drug effect analysis. All samples were sequenced using Illumina HiSeq 4000 and the microbial composition was determined.Results. Four species were enriched in the patients with AS: Flavonifractor plautii, Oscillibacter, Parabacteroides distasonis and Bacteroides nordii (HC vs. NM, P<0.05); only F. plautii was found to be significantly changed in the NM-HC comparison. No additional species were found in the HC vs. CHM analysis, which indicated a beneficial effect of CHM in removing the other three strains. F. plautii was found to be significantly increased in the comparison between the HC and WM groups, along with four other species (Clostridium bolteae, Clostridiales bacterium 1_7_47FAA, C. asparagiforme and C. hathewayi). The patients with AS harboured more bacterial species associated with carbohydrate metabolism and glycan biosynthesis in their faeces. They also had bacterial profiles less able to biodegrade xenobiotics or synthesize and transport vitamins.Conclusion. The gut microbiota of the patients with AS varied from that of the HCs, and the treatment had an impact on this divergence. Our data provide insight that could guide improvements in AS treatment.
Collapse
Affiliation(s)
- RunYue Huang
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120 or 510006, PR China
| | - Fang Li
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120 or 510006, PR China
| | - Yingyan Zhou
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120 or 510006, PR China
- Postdoctoral Mobile Research Station, Guangzhou 510006, PR China
| | - Zhenhua Zeng
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120 or 510006, PR China
| | - Xiaohong He
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120 or 510006, PR China
| | - Lihua Fang
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120 or 510006, PR China
| | - Feng Pan
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120 or 510006, PR China
| | - Yile Chen
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120 or 510006, PR China
| | - Jiehua Lin
- Nephropathy Center, The Affiliated Jiangmen TCM Hospital, Jinan University, Jiangmen 529000, PR China
| | - Jie Li
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120 or 510006, PR China
| | - Dongni Qiu
- Department of Traditional Chinese Medicine, Guangdong Armed Police Corps Hospital, Guangzhou 510507, PR China
| | - Yinping Tian
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120 or 510006, PR China
| | - Xi Tan
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120 or 510006, PR China
| | - Yanni Song
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120 or 510006, PR China
| | - Yongyue Xu
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120 or 510006, PR China
| | - Yonghui Lai
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120 or 510006, PR China
| | - Hao Yi
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120 or 510006, PR China
| | - Qiang Gao
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120 or 510006, PR China
| | - Xiaodong Fang
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120 or 510006, PR China
| | - Mingming Shi
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120 or 510006, PR China
| | - Chu Zhou
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120 or 510006, PR China
| | - Jinqun Huang
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120 or 510006, PR China
| | - Yi-Ting He
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120 or 510006, PR China
| |
Collapse
|
15
|
Zhang F, Ma C, Zhang B, Bi L. Dynamic changes in gut microbiota under the influence of smoking and TNF-α-blocker in patients with ankylosing spondylitis. Clin Rheumatol 2020; 39:2653-2661. [PMID: 32219620 DOI: 10.1007/s10067-020-05032-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/21/2020] [Accepted: 03/05/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVES This study aimed to investigate the relationship among smoking, TNF-α-blocker therapy, and the dynamic changes in gut microbiota in patients with ankylosing spondylitis (AS). METHODS Using a 16S rRNA sequence, 98 fecal samples of 20 AS patients collected after 0, 1, 3 and 6 months of anti-TNF-α treatment and from 20 matched health controls were examined. The variation in composition, abundance, and diversity of gut microbiota was analyzed. The dynamic effects of smoking and treatment on gut microbiota and therapeutic efficacy in AS patients were studied. RESULTS The increased relative abundance of microbiota in AS nonsmokers was g_Comamonas and g_Desulfovibrio, while that in AS smokers was g_Actinomyces, g_Collinsella, g_Lachnospiraceae_UCG-008, and g_Paraprevotella. The relative abundance of gut microbiota showed dynamic variation. The improvement rate of ASDAS in AS nonsmokers was higher than that in AS smokers (2.297 vs 1.736) after anti-TNF-α treatment. The β-diversity of gut microbiota in AS smokers was lower than that in AS nonsmokers and improved with treatment. CONCLUSIONS Both smoking and TNF-α-blocker had significant effects on the composition, relative abundance, and diversity of gut microbiota in AS patients. The AS smokers characteristically shared g_Collinsella and g_Dorea. The relative abundance of gut microbiota revealed high variability and was in dynamic fluctuation during treatment. The response of gut microbiota to anti-TNF-α treatment was found to be heterogeneous and selective. AS nonsmokers showed a greater improvement rate of ASDAS-CRP with treatment than AS smokers did. The AS smokers showed a lower β-diversity of gut microbiota, and improved after treatment. Key Points • Characterized the dynamic variation in gut microbiota in AS patients classified as smokers and nonsmokers during treatment with anti-TNF-α. • Confirmed the interaction between smoking, anti-TNF-α therapy, and gut microbiota.
Collapse
Affiliation(s)
- Fangze Zhang
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, No.126 Xiantai Street, Changchun, 130033, Jilin, People's Republic of China
| | - Cuili Ma
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, No.126 Xiantai Street, Changchun, 130033, Jilin, People's Republic of China
| | - Bin Zhang
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, No.126 Xiantai Street, Changchun, 130033, Jilin, People's Republic of China.
| | - Liqi Bi
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, No.126 Xiantai Street, Changchun, 130033, Jilin, People's Republic of China.
| |
Collapse
|
16
|
Abstract
The etiology of primary sclerosing cholangitis (PSC) is unknown. I present a case which may be indicative of a causal link between Bartonella infection and PSC. The patient presented with complaints of abdominal pain and bloody diarrhea. A colonoscopy demonstrated chronic inflammation and changes consistent with ulcerative colitis. Routine laboratory studies revealed elevated liver function tests (LFTs); ultrasound and magnetic resonance imaging (MRI) confirmed the diagnosis of PSC. Bartonella serology was positive. It is established that Bartonella infection is associated with both gastrointestinal inflammation and autoimmunity; indeed, there is an animal model for Bartonella-induced PSC. Bartonella is susceptible to treatment with vancomycin and there are case reports and small series that demonstrate that PSC responds to treatment with oral vancomycin. Because of this, it is postulated that at least some cases of PSC may be associated with Bartonella infection. The patient in this report was treated with oral vancomycin and, since then, has been in remission for both colitis and PSC. Since vancomycin is not systemically absorbed, the premise is that he suffered from Bartonella colitis and an autoimmune reaction to Bartonella causing PSC. This premise warrants further study.
Collapse
|
17
|
Frasca L, Lande R. Toll-like receptors in mediating pathogenesis in systemic sclerosis. Clin Exp Immunol 2020; 201:14-24. [PMID: 32048277 DOI: 10.1111/cei.13426] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2020] [Indexed: 02/06/2023] Open
Abstract
Toll-like receptors (TLRs) are evolutionarily conserved receptors essential for the host defence against pathogens. Both immune and non-immune cells can express TLRs, although at different levels. Systemic sclerosis (SSc) is a chronic disease in which autoimmunity, dysregulated profibrotic mediator release and activation of fibroblasts lead to dysregulated collagen deposition and fibrosis. There is now increasing knowledge that the innate immune system and, in particular, TLRs take a part in SSc pathogenesis. The list of endogenous ligands that can stimulate TLRs in SSc is growing: these ligands represent specific danger-associated molecular patterns (DAMPs), involved either in the initiation or the perpetuation of inflammation, and in the release of factors that sustain the fibrotic process or directly stimulate the cells that produce collagen and the endothelial cells. This review reports evidences concerning TLR signalling involvement in SSc. We report the new DAMPs, as well as the TLR-linked pathways involved in disease, with emphasis on type I interferon signature in SSc, the role of plasmacytoid dendritic cells (pDCs) and platelets. The dissection of the contribution of all these pathways to disease, and their correlation with the disease status, as well as their values as prognostic tools, can help to plan timely intervention and design new drugs for more appropriate therapeutic strategies.
Collapse
Affiliation(s)
- L Frasca
- National Centre for Drug Research and Evaluation, Pharmacological Research and Experimental Therapy Unit, Istituto Superiore di Sanità, Rome, Italy
| | - R Lande
- National Centre for Drug Research and Evaluation, Pharmacological Research and Experimental Therapy Unit, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
18
|
Pan H, Guo R, Ju Y, Wang Q, Zhu J, Xie Y, Zheng Y, Li T, Liu Z, Lu L, Li F, Tong B, Xiao L, Xu X, Leung ELH, Li R, Yang H, Wang J, Zhou H, Jia H, Liu L. A single bacterium restores the microbiome dysbiosis to protect bones from destruction in a rat model of rheumatoid arthritis. MICROBIOME 2019; 7:107. [PMID: 31315667 PMCID: PMC6637628 DOI: 10.1186/s40168-019-0719-1] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 07/03/2019] [Indexed: 05/04/2023]
Abstract
Background Early treatment is key for optimizing the therapeutic success of drugs, and the current initiating treatment that blocks the progression of bone destruction during the pre-arthritic stages remains unsatisfactory. The microbial disorder in rheumatoid arthritis (RA) patients is significantly reversed with effective treatment. Modulating aberrant gut microbiomes into a healthy state is a potential therapeutic approach for preventing bone damage. Results By using metagenomic shotgun sequencing and a metagenome-wide association study, we assessed the effect of Lactobacillus casei (L. casei) on the induction of arthritis as well as on the associated gut microbiota and immune disorders in adjuvant-induced arthritis (AIA) rats. Treatment of AIA rats with L. casei inhibited joint swelling, lowered arthritis scores, and prevented bone destruction. Along with the relief of arthritis symptoms, dysbiosis in the microbiome of arthritic rats was significantly reduced after L. casei intervention. The relative abundance of AIA-decreased Lactobacillus strains, including Lactobacillus hominis, Lactobacillus reuteri, and Lactobacillus vaginalis, were restored to normal and Lactobacillus acidophilus was upregulated by the administration of L. casei to the AIA rats. Moreover, L. casei downregulated the expression of pro-inflammatory cytokines, which are closely linked to the effect of the L. casei treatment-associated microbes. Functionally, the maintenance of the redox balance of oxidative stress was involved in the improvement in the L. casei-treated AIA rats. Conclusion A single bacterium, L. casei (ATCC334), was able to significantly suppress the induction of AIA and protect bones from destruction in AIA rats by restoring the microbiome dysbiosis in the gut, indicating that using probiotics may be a promising strategy for treating RA, especially in the early stage of the disease. Electronic supplementary material The online version of this article (10.1186/s40168-019-0719-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hudan Pan
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macao University of Science and Technology, Macao, China
| | - Ruijin Guo
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macao University of Science and Technology, Macao, China
- BGI-Shenzhen, Shenzhen, 518083 China
- China National Genebank, BGI-Shenzhen, Shenzhen, 518120 China
| | - Yanmei Ju
- BGI-Shenzhen, Shenzhen, 518083 China
- China National Genebank, BGI-Shenzhen, Shenzhen, 518120 China
| | - Qi Wang
- BGI-Shenzhen, Shenzhen, 518083 China
- China National Genebank, BGI-Shenzhen, Shenzhen, 518120 China
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083 China
| | - Jie Zhu
- BGI-Shenzhen, Shenzhen, 518083 China
- China National Genebank, BGI-Shenzhen, Shenzhen, 518120 China
| | - Ying Xie
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macao University of Science and Technology, Macao, China
| | - Yanfang Zheng
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macao University of Science and Technology, Macao, China
- Fujian University of Traditional Chinese Medicine, No.1, Qiuyang Road, Minhoushangjie, Fuzhou, 350122 Fujian China
| | - Ting Li
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macao University of Science and Technology, Macao, China
| | - Zhongqiu Liu
- International Institute for Translational Research of Traditional Chinese Medicine of Guangzhou University of Chinese Medicine, Guangzhou, 510006 Guangdong China
| | - Linlin Lu
- International Institute for Translational Research of Traditional Chinese Medicine of Guangzhou University of Chinese Medicine, Guangzhou, 510006 Guangdong China
| | - Fei Li
- BGI-Shenzhen, Shenzhen, 518083 China
- China National Genebank, BGI-Shenzhen, Shenzhen, 518120 China
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083 China
| | - Bin Tong
- BGI-Shenzhen, Shenzhen, 518083 China
- China National Genebank, BGI-Shenzhen, Shenzhen, 518120 China
| | - Liang Xiao
- BGI-Shenzhen, Shenzhen, 518083 China
- China National Genebank, BGI-Shenzhen, Shenzhen, 518120 China
- Shenzhen Engineering Laboratory of Detection and Intervention of human intestinal microbiome, BGI-Shenzhen, Shenzhen, 518083 China
| | - Xun Xu
- BGI-Shenzhen, Shenzhen, 518083 China
- China National Genebank, BGI-Shenzhen, Shenzhen, 518120 China
| | - Elaine Lai-Han Leung
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macao University of Science and Technology, Macao, China
| | - Runze Li
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macao University of Science and Technology, Macao, China
| | - Huanming Yang
- BGI-Shenzhen, Shenzhen, 518083 China
- China National Genebank, BGI-Shenzhen, Shenzhen, 518120 China
- James D. Watson Institute of Genome Sciences, Hangzhou, 310058 China
| | - Jian Wang
- BGI-Shenzhen, Shenzhen, 518083 China
- China National Genebank, BGI-Shenzhen, Shenzhen, 518120 China
- James D. Watson Institute of Genome Sciences, Hangzhou, 310058 China
| | - Hua Zhou
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macao University of Science and Technology, Macao, China
| | - Huijue Jia
- BGI-Shenzhen, Shenzhen, 518083 China
- China National Genebank, BGI-Shenzhen, Shenzhen, 518120 China
| | - Liang Liu
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macao University of Science and Technology, Macao, China
| |
Collapse
|
19
|
Vivek Aithal PR, Akshai Shetty KR, Dinesh MR, Amarnath BC, Prashanth CS, Roopak MD. In vitro evaluation of microbial contamination and the disinfecting efficacy of chlorhexidine on orthodontic brackets. Prog Orthod 2019; 20:17. [PMID: 31041551 PMCID: PMC6491528 DOI: 10.1186/s40510-019-0270-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 03/19/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Contamination of orthodontic appliances is due to the unhygienic practices followed during manufacturing and packaging processes, which may lead to cross-contamination. Although literature has indicated the need for sterilization or disinfection of orthodontic appliances before using in the oral cavity, this is still not employed in routine clinical practice. In this view, the current study evaluates the bacterial load on orthodontic brackets along with the disinfecting efficacy of chlorhexidine. METHODS A total of 140 brackets were obtained from four different manufacturers and divided into six groups: group 1 (American Orthodontics; n = 30), group 2 (3M Unitek; n = 30), group 3 (Ortho Organizers; n = 30), group 4 (China Dental Orthodontic; n = 30), group 5 (negative control; n = 10), and group 6 (positive control; n = 10). Various microbiological and biochemical tests were conducted on the brackets to detect the type and growth of bacteria. Brackets that showed microbial contamination were then subjected to disinfection using 0.01% and 2% chlorhexidine solutions. RESULTS Microbial contamination was detected on brackets of all the four groups. Bacteria, including Staphylococcus aureus, S. epidermidis, Lactobacilli, Klebsiella pneumoniae, Bacillus licheniformis, and B. cereus, were identified in these groups. Upon disinfection with 0.01% chlorhexidine solution, brackets in group 2 displayed complete decontamination, while all brackets in the other groups containing gram-negative bacteria exhibited complete decontamination with 2% chlorhexidine. CONCLUSION Orthodontic brackets received from four manufacturers showed high bacterial contamination. Disinfecting ability of 2% chlorhexidine proved highly effective in destroying both gram-positive and gram-negative bacteria. Therefore, use of 2% chlorhexidine in clinical practice for the disinfection of orthodontic brackets is suggested, before placement in the oral cavity.
Collapse
Affiliation(s)
| | | | - M R Dinesh
- DAPM RV Dental College, Bengaluru, Karnataka, 560078, India
| | - B C Amarnath
- DAPM RV Dental College, Bengaluru, Karnataka, 560078, India
| | - C S Prashanth
- DAPM RV Dental College, Bengaluru, Karnataka, 560078, India
| | | |
Collapse
|
20
|
Glassner K, Quigley EM, Franco L, Victor DW. Autoimmune liver disease and the enteric microbiome. AIMS Microbiol 2018; 4:334-346. [PMID: 31294219 PMCID: PMC6604930 DOI: 10.3934/microbiol.2018.2.334] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 05/08/2018] [Indexed: 12/13/2022] Open
Abstract
The human enteric microbiome is highly complex and has more than 150 times more genes within it than its host. The host and the microbiome have a commensurate relationship that can evolve over time. The typically symbiotic relationship between the two can become pathogenic. The microbiome composition in adults reflects their history of exposure to bacteria and environmental factors during early life, their genetic background, age, interactions with the immune system, geographical location, and, most especially, their diet. Similarly, these factors are thought to contribute to the development of autoimmune disease. It is possible that alterations in the intestinal microbiome could lead to liver disease. There is emerging data for the contribution of the microbiome in development of primary sclerosing cholangitis, primary biliary cholangitis, and autoimmune hepatitis; liver disorders associated with aberrant immune function in genetically susceptible individuals.
Collapse
Affiliation(s)
- Kerri Glassner
- Lynda K and David M Underwood Center for Digestive Disorders, Division of Gastroenterology and Hepatology, Houston Methodist Hospital, Weill Cornell Medical College, 6550 Fannin Street, SM 1201, Houston, TX 77030, USA
| | - Eamonn Mm Quigley
- Lynda K and David M Underwood Center for Digestive Disorders, Division of Gastroenterology and Hepatology, Houston Methodist Hospital, Weill Cornell Medical College, 6550 Fannin Street, SM 1201, Houston, TX 77030, USA
| | - Lissa Franco
- Lynda K and David M Underwood Center for Digestive Disorders, Division of Gastroenterology and Hepatology, Houston Methodist Hospital, Weill Cornell Medical College, 6550 Fannin Street, SM 1201, Houston, TX 77030, USA
| | - David W Victor
- Lynda K and David M Underwood Center for Digestive Disorders, Division of Gastroenterology and Hepatology, Houston Methodist Hospital, Weill Cornell Medical College, 6550 Fannin Street, SM 1201, Houston, TX 77030, USA.,Sherrie and Alan Conover Center for Liver Disease, Houston Methodist Hospital, Weill Cornell Medical College, 6550 Fannin Street, SM 1201, Houston, TX 77030, USA
| |
Collapse
|
21
|
Terato K, Waritani T, Fukai R, Shionoya H, Itoh H, Katayama K. Contribution of bacterial pathogens to evoking serological disease markers and aggravating disease activity in rheumatoid arthritis. PLoS One 2018; 13:e0190588. [PMID: 29408886 PMCID: PMC5800560 DOI: 10.1371/journal.pone.0190588] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 12/18/2017] [Indexed: 12/24/2022] Open
Abstract
Commensal bacteria and their pathogenic components in the gastrointestinal tract and oral cavity may play pathological roles in autoimmune diseases. To study the possible involvement of bacterial pathogens in autoimmune diseases, IgG and IgA antibodies against pathogenic components produced by three strains of commensal bacteria, Escherichia coli-lipopolysaccharide (E. coli-LPS), Porphyromonas gingivalis-LPS (Pg-LPS) and peptidoglycan polysaccharide (PG-PS) from Streptococcus pyogenes, were determined by an improved ELISA system for sera from two groups of patients with rheumatoid arthritis (RA), who met rapid radiographic progression (RRP) criteria and non-RRP, and compared to normal (NL) controls. Antibody responses to these bacterial pathogens are unique and consistent in individuals, and no fundamental difference was observed between RA and NL controls. Despite the similar antibody responses to pathogens, lower IgG or higher IgA and consequent higher IgA/IgG antibody ratio among the patients with RA related to disease marker levels and disease activity. Peculiarly, the IgA/IgG anti-Pg-LPS antibody ratio resulted from lower IgG and higher IgA antibody responses to Pg-LPS strongly correlated not only with rheumatoid factor (RF), but also correlated with erythrocyte sedimentation rate (ESR), C-reactive protein (CRP) and disease activity score of 28 joints with ESR (DAS28-ESR) in the RRP group. In contrast, the IgA/IgG anti-E. coli-LPS and anti-PG-PS antibody ratio correlated or tended to correlate with RF, ESR, CRP, and DAS28-ESR in the non-RRP group, whereas either the IgG or IgA anti-Pg-LPS antibody levels and consequent IgA/IgG anti-Pg-LPS antibody ratio did not correlate with any clinical marker levels in this group. Notably, anti-circular-citrullinated peptide (CCP) antibody levels, which did not correlate with either IgG or IgA antibody levels to any pathogens, did not correlate with severity of arthritis in both RRP and non-RRP. Taken together, we propose that multiple environmental pathogens, which overwhelm the host antibody defense function, contribute independently or concomitantly to evoking disease makers and aggravating disease activity, and affect disease outcomes. TRIAL REGISTRATION UMIN CTR UMIN000012200.
Collapse
Affiliation(s)
- Kuniaki Terato
- Department of Research and Development, Chondrex Inc. Redmond, WA, United States of America
| | - Takaki Waritani
- Department of Research and Development, Chondrex Inc. Redmond, WA, United States of America
| | | | - Hiroshi Shionoya
- Research Lab Section 5, Asama Chemicals Inc. Chiyoda, Tokyo, Japan
| | - Hiroshi Itoh
- Department of Orthopedic Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Kou Katayama
- Katayama Orthopedic Rheumatology Clinic, Asahikawa, Hokkaido, Japan
| |
Collapse
|
22
|
Wen C, Zheng Z, Shao T, Liu L, Xie Z, Le Chatelier E, He Z, Zhong W, Fan Y, Zhang L, Li H, Wu C, Hu C, Xu Q, Zhou J, Cai S, Wang D, Huang Y, Breban M, Qin N, Ehrlich SD. Quantitative metagenomics reveals unique gut microbiome biomarkers in ankylosing spondylitis. Genome Biol 2017; 18:142. [PMID: 28750650 PMCID: PMC5530561 DOI: 10.1186/s13059-017-1271-6] [Citation(s) in RCA: 228] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 06/27/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The assessment and characterization of the gut microbiome has become a focus of research in the area of human autoimmune diseases. Ankylosing spondylitis is an inflammatory autoimmune disease and evidence showed that ankylosing spondylitis may be a microbiome-driven disease. RESULTS To investigate the relationship between the gut microbiome and ankylosing spondylitis, a quantitative metagenomics study based on deep shotgun sequencing was performed, using gut microbial DNA from 211 Chinese individuals. A total of 23,709 genes and 12 metagenomic species were shown to be differentially abundant between ankylosing spondylitis patients and healthy controls. Patients were characterized by a form of gut microbial dysbiosis that is more prominent than previously reported cases with inflammatory bowel disease. Specifically, the ankylosing spondylitis patients demonstrated increases in the abundance of Prevotella melaninogenica, Prevotella copri, and Prevotella sp. C561 and decreases in Bacteroides spp. It is noteworthy that the Bifidobacterium genus, which is commonly used in probiotics, accumulated in the ankylosing spondylitis patients. Diagnostic algorithms were established using a subset of these gut microbial biomarkers. CONCLUSIONS Alterations of the gut microbiome are associated with development of ankylosing spondylitis. Our data suggest biomarkers identified in this study might participate in the pathogenesis or development process of ankylosing spondylitis, providing new leads for the development of new diagnostic tools and potential treatments.
Collapse
Affiliation(s)
- Chengping Wen
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Zhijun Zheng
- Realbio Genomics Institute, Shanghai, 200123, China
| | - Tiejuan Shao
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Lin Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Department of Infectious Diseases, the First Affiliated College of Medicine, Zhejiang University, Hangzhou, 310003, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, 310003, China
| | - Zhijun Xie
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Emmanuelle Le Chatelier
- INRA, Institut National de la Recherche Agronomique, Metagenopolis, Jouy en Josas, 78350, France
| | - Zhixing He
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Wendi Zhong
- Realbio Genomics Institute, Shanghai, 200123, China
| | - Yongsheng Fan
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | | | - Haichang Li
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Chunyan Wu
- Realbio Genomics Institute, Shanghai, 200123, China
| | - Changfeng Hu
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Qian Xu
- Realbio Genomics Institute, Shanghai, 200123, China
| | - Jia Zhou
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Shunfeng Cai
- Realbio Genomics Institute, Shanghai, 200123, China
| | - Dawei Wang
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yun Huang
- Realbio Genomics Institute, Shanghai, 200123, China
| | - Maxime Breban
- Rheumatology Division, Ambroise-Paré Hospital, AP-HP, 9, avenue Charles-de-Gaulle, 92100, Boulogne-Billancourt, France
| | - Nan Qin
- Realbio Genomics Institute, Shanghai, 200123, China. .,State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Department of Infectious Diseases, the First Affiliated College of Medicine, Zhejiang University, Hangzhou, 310003, China. .,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, 310003, China.
| | - Stanislav Dusko Ehrlich
- INRA, Institut National de la Recherche Agronomique, Metagenopolis, Jouy en Josas, 78350, France. .,King's College London, Centre for Host-Microbiome Interactions, Dental Institute Central Office, Guy's Hospital, London Bridge, London, SE1 9RT, UK.
| |
Collapse
|
23
|
Aranda-Uribe IS, Ortega E, Martínez-Cordero E. Immunization of BALB/c mice with pigeon IgY induces the production of anti-IgG autoantibodies. Autoimmunity 2017; 50:336-345. [PMID: 28699799 DOI: 10.1080/08916934.2017.1344974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The breakdown of immunological tolerance due to the activation of autoreactive B and T cells triggers physiopathological processes. An example of such conditions is the production of IgG autoantibodies specific for the Fc portion of IgG (anti-Fcγ IgG). Previous reports have shown that patients with pigeon-related hypersensitivity pneumonitis exhibit an increase in the serum levels of anti-Fcγ IgG. There is no in vivo model for the study of this condition and the immunological mechanisms of tolerance breakdown associated with sensitization by pigeon antigens are still unknown. In this work, we show that the repeated immunization of BALB/c mice with pigeon IgY during 16-weeks induces the production of anti-Fcγ IgG and keeps their high levels for seven weeks. The late appearance of anti-Fcγ IgG autoantibodies in the plasma is similar to what has been reported in other experimental autoimmune models. With the occurrence of anti-Fcγ IgG, there is a reduction in the proportion of Foxp3 + cells (regulatory T cells, Tregs) within the population of splenic CD4 + CD25 + T cells. Thus, our data showed that the immunization of BALB/c mice with IgY promotes the production of anti-Fcγ IgG along with a decrease in Tregs in the spleen. We propose that immunization of mice with pigeon antigens, like IgY can provide a model to study the immunological mechanisms involved in the development of pigeon-related hypersensitivity pneumonitis.
Collapse
Affiliation(s)
- Ivan Sammir Aranda-Uribe
- a Facultad de Medicina , Universidad Nacional Autónoma de México, Posgrado Ciencias Biológicas , México City , México.,b Laboratorio de Autoinmunidad , Unidad de Investigación INER , México City , México
| | - Enrique Ortega
- c Instituto de Investigaciones Biomédicas , Universidad Nacional Autónoma de México , México City , México
| | | |
Collapse
|
24
|
Viladomiu M, Kivolowitz C, Abdulhamid A, Dogan B, Victorio D, Castellanos JG, Woo V, Teng F, Tran NL, Sczesnak A, Chai C, Kim M, Diehl GE, Ajami NJ, Petrosino JF, Zhou XK, Schwartzman S, Mandl LA, Abramowitz M, Jacob V, Bosworth B, Steinlauf A, Scherl EJ, Wu HJJ, Simpson KW, Longman RS. IgA-coated E. coli enriched in Crohn's disease spondyloarthritis promote T H17-dependent inflammation. Sci Transl Med 2017; 9:eaaf9655. [PMID: 28179509 PMCID: PMC6159892 DOI: 10.1126/scitranslmed.aaf9655] [Citation(s) in RCA: 223] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 08/12/2016] [Accepted: 12/27/2016] [Indexed: 12/21/2022]
Abstract
Peripheral spondyloarthritis (SpA) is a common extraintestinal manifestation in patients with active inflammatory bowel disease (IBD) characterized by inflammatory enthesitis, dactylitis, or synovitis of nonaxial joints. However, a mechanistic understanding of the link between intestinal inflammation and SpA has yet to emerge. We evaluated and functionally characterized the fecal microbiome of IBD patients with or without peripheral SpA. Coupling the sorting of immunoglobulin A (IgA)-coated microbiota with 16S ribosomal RNA-based analysis (IgA-seq) revealed a selective enrichment in IgA-coated Escherichia coli in patients with Crohn's disease-associated SpA (CD-SpA) compared to CD alone. E. coli isolates from CD-SpA-derived IgA-coated bacteria were similar in genotype and phenotype to an adherent-invasive E. coli (AIEC) pathotype. In comparison to non-AIEC E. coli, colonization of germ-free mice with CD-SpA E. coli isolates induced T helper 17 cell (TH17) mucosal immunity, which required the virulence-associated metabolic enzyme propanediol dehydratase (pduC). Modeling the increase in mucosal and systemic TH17 immunity we observed in CD-SpA patients, colonization of interleukin-10-deficient or K/BxN mice with CD-SpA-derived E. coli lead to more severe colitis or inflammatory arthritis, respectively. Collectively, these data reveal the power of IgA-seq to identify immunoreactive resident pathosymbionts that link mucosal and systemic TH17-dependent inflammation and offer microbial and immunophenotype stratification of CD-SpA that may guide medical and biologic therapy.
Collapse
Affiliation(s)
- Monica Viladomiu
- Jill Roberts Institute for Research in Inflammatory Bowel Disease (IBD), Weill Cornell Medicine, New York, NY 10021, USA
| | - Charles Kivolowitz
- Jill Roberts Institute for Research in Inflammatory Bowel Disease (IBD), Weill Cornell Medicine, New York, NY 10021, USA
| | - Ahmed Abdulhamid
- Jill Roberts Institute for Research in Inflammatory Bowel Disease (IBD), Weill Cornell Medicine, New York, NY 10021, USA
| | - Belgin Dogan
- College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Daniel Victorio
- Jill Roberts Institute for Research in Inflammatory Bowel Disease (IBD), Weill Cornell Medicine, New York, NY 10021, USA
| | - Jim G Castellanos
- Jill Roberts Institute for Research in Inflammatory Bowel Disease (IBD), Weill Cornell Medicine, New York, NY 10021, USA
| | - Viola Woo
- Jill Roberts Institute for Research in Inflammatory Bowel Disease (IBD), Weill Cornell Medicine, New York, NY 10021, USA
| | - Fei Teng
- Department of Immunobiology, University of Arizona, Tucson, AZ 85719, USA
| | - Nhan L Tran
- Department of Immunobiology, University of Arizona, Tucson, AZ 85719, USA
| | - Andrew Sczesnak
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94709, USA
| | - Christina Chai
- Jill Roberts Institute for Research in Inflammatory Bowel Disease (IBD), Weill Cornell Medicine, New York, NY 10021, USA
| | - Myunghoo Kim
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Gretchen E Diehl
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nadim J Ajami
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Joseph F Petrosino
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xi K Zhou
- Division of Biostatistics and Epidemiology, Weill Cornell Medical College, New York, NY 10065, USA
| | | | - Lisa A Mandl
- Hospital for Special Surgery, New York, NY 10021, USA
| | - Meira Abramowitz
- Jill Roberts Center for IBD, Weill Cornell Medicine, New York, NY 10021, USA
| | - Vinita Jacob
- Jill Roberts Center for IBD, Weill Cornell Medicine, New York, NY 10021, USA
| | - Brian Bosworth
- Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Adam Steinlauf
- Jill Roberts Center for IBD, Weill Cornell Medicine, New York, NY 10021, USA
| | - Ellen J Scherl
- Jill Roberts Center for IBD, Weill Cornell Medicine, New York, NY 10021, USA
| | - Hsin-Jung Joyce Wu
- Department of Immunobiology, University of Arizona, Tucson, AZ 85719, USA
| | - Kenneth W Simpson
- College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Randy S Longman
- Jill Roberts Institute for Research in Inflammatory Bowel Disease (IBD), Weill Cornell Medicine, New York, NY 10021, USA.
- Jill Roberts Center for IBD, Weill Cornell Medicine, New York, NY 10021, USA
| |
Collapse
|
25
|
Rogers MB, Aveson V, Firek B, Yeh A, Brooks B, Brower-Sinning R, Steve J, Banfield JF, Zureikat A, Hogg M, Boone BA, Zeh HJ, Morowitz MJ. Disturbances of the Perioperative Microbiome Across Multiple Body Sites in Patients Undergoing Pancreaticoduodenectomy. Pancreas 2017; 46:260-267. [PMID: 27846140 PMCID: PMC5235958 DOI: 10.1097/mpa.0000000000000726] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The goals of this study were to characterize bacterial communities within fecal samples, pancreatic fluid, bile, and jejunal contents from patients undergoing pancreaticoduodenectomy (PD) and to identify associations between microbiome profiles and clinical variables. METHODS Fluid was collected from the pancreas, common bile duct, and proximal jejunum from 50 PD patients. Postoperative fecal samples were also collected. The microbial burden within samples was quantified with droplet digital polymerase chain reaction. Bacterial 16S ribosomal RNA gene sequences were amplified, sequenced, and analyzed. Data from fecal samples were compared with publicly available data obtained from volunteers. RESULTS Droplet digital polymerase chain reaction confirmed the presence of bacteria in all sample types, including pancreatic fluid. Relative to samples from the American Gut Project, fecal samples from PD patients were enriched with Klebsiella and Bacteroides and were depleted of anaerobic taxa (eg, Roseburia and Faecalibacterium). Similar patterns were observed within PD pancreas, bile, and jejunal samples. Postoperative fecal samples from patients with a pancreatic fistula contained increased abundance of Klebsiella and decreased abundance of commensal anaerobes, for example, Ruminococcus. CONCLUSIONS This study confirms the presence of altered bacterial populations within samples from PD patients. Future research must validate these findings and may evaluate targeted microbiome modifications to improve outcomes in PD patients.
Collapse
Affiliation(s)
| | - Victoria Aveson
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Brian Firek
- Division of Pediatric General and Thoracic Surgery, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, PA
| | - Andrew Yeh
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Brandon Brooks
- Department of Earth and Planetary Sciences, University of California Berkeley, Berkeley, CA
| | | | - Jennifer Steve
- Division of GI Surgical Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Jillian F. Banfield
- Department of Earth and Planetary Sciences, University of California Berkeley, Berkeley, CA
| | - Amer Zureikat
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA,Division of GI Surgical Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Melissa Hogg
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA,Division of GI Surgical Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Brian A. Boone
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA,Division of GI Surgical Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Herbert J. Zeh
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA,Division of GI Surgical Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Michael J. Morowitz
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA,Division of Pediatric General and Thoracic Surgery, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, PA,Address Correspondence to: Michael J. Morowitz, MD, Division of Pediatric Surgery, Children’s Hospital of Pittsburgh of UPMC, 4401 Penn Avenue, 7th Floor Faculty Pavilion, Pittsburgh, PA 15244, , phone 412-692-7282, fax 412-692-8299
| |
Collapse
|
26
|
Guarneri F, Giuffrida R, Di Bari F, Cannavò SP, Benvenga S. Thyroid Autoimmunity and Lichen. Front Endocrinol (Lausanne) 2017; 8:146. [PMID: 28701998 PMCID: PMC5484774 DOI: 10.3389/fendo.2017.00146] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 06/09/2017] [Indexed: 12/12/2022] Open
Abstract
Lichen planus (LP) and lichen sclerosus (LS) are cutaneous-mucous diseases with uncertain epidemiology. Current data, which are likely to be underestimated, suggest a prevalence in the general population of 0.1-4% for cutaneous LP, 1.27-2.0% for oral LP, and 0.1-3.3% for LS. While etiology of lichen is still unknown, clinical and histological evidence show an (auto)immune pathogenesis. Association of lichen with autoimmune thyroid disease (AITD) has been investigated in few studies. This association appears better defined in the case of LS, while is more controversial for LP. In both situations, the frequency of the association is higher in females. We review the available literature on the correlation between the different types of lichen and AITD, and the literature on the genetic risk factors which are shared by both conditions. Such data suggest that a common pathogenic mechanism could be the cause for co-occurrence of lichen and AITD, at least in some patients. Additionally, analyzing literature data and in continuity with our previous work on other autoimmune diseases, we suggest that molecular mimicry could trigger both diseases, and thus explain their co-occurrence.
Collapse
Affiliation(s)
- Fabrizio Guarneri
- Department of Clinical and Experimental Medicine – Dermatology, University of Messina, Messina, Italy
- *Correspondence: Fabrizio Guarneri,
| | - Roberta Giuffrida
- Department of Clinical and Experimental Medicine – Dermatology, University of Messina, Messina, Italy
| | - Flavia Di Bari
- Department of Clinical and Experimental Medicine – Endocrinology, University of Messina, Messina, Italy
| | | | - Salvatore Benvenga
- Department of Clinical and Experimental Medicine – Endocrinology, University of Messina, Messina, Italy
- Master Program on Childhood, Adolescent and Women’s Endocrine Health, University of Messina, Messina, Italy
- Interdepartmental Program of Molecular & Clinical Endocrinology and Women’s Endocrine Health, University Hospital Policlinico “G. Martino”, Messina, Italy
| |
Collapse
|
27
|
Yang L, Wang L, Wang X, Xian CJ, Lu H. A Possible Role of Intestinal Microbiota in the Pathogenesis of Ankylosing Spondylitis. Int J Mol Sci 2016; 17:ijms17122126. [PMID: 27999312 PMCID: PMC5187926 DOI: 10.3390/ijms17122126] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/09/2016] [Accepted: 12/14/2016] [Indexed: 12/12/2022] Open
Abstract
Ankylosing spondylitis (AS) is a chronic inflammatory disease primarily affecting the sacroiliac joints and the spine, for which the pathogenesis is thought to be a result of the combination of host genetic factors and environmental triggers. However, the precise factors that determine one’s susceptibility to AS remain to be unraveled. With 100 trillion bacteria residing in the mammalian gut having established a symbiotic relation with their host influencing many aspects of host metabolism, physiology, and immunity, a growing body of evidence suggests that intestinal microbiota may play an important role in AS. Several mechanisms have been suggested to explain the potential role of the microbiome in the etiology of AS, such as alterations of intestinal permeability, stimulation of immune responses, and molecular mimicry. In this review, the existing evidence for the involvement of the microbiome in AS pathogenesis was discussed and the potential of intestinal microbiome-targeting strategies in the prevention and treatment of AS was evaluated.
Collapse
Affiliation(s)
- Lianjun Yang
- Academy of Orthopedics of Guangdong Province, Orthopaedic Hospital of Guangdong Province, Department of Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China.
| | - Liping Wang
- Academy of Orthopedics of Guangdong Province, Orthopaedic Hospital of Guangdong Province, Department of Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China.
- Sansom Institute for Health Research and School of Pharmacy and Medical Sciences, University of South Australia, Adelaide SA5001, Australia.
| | - Xin Wang
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane QLD4059, Australia.
| | - Cory J Xian
- Academy of Orthopedics of Guangdong Province, Orthopaedic Hospital of Guangdong Province, Department of Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China.
- Sansom Institute for Health Research and School of Pharmacy and Medical Sciences, University of South Australia, Adelaide SA5001, Australia.
| | - Hai Lu
- Academy of Orthopedics of Guangdong Province, Orthopaedic Hospital of Guangdong Province, Department of Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China.
| |
Collapse
|
28
|
Christopoulos G, Christopoulou V, Routsias JG, Babionitakis A, Antoniadis C, Vaiopoulos G. Greek rheumatoid arthritis patients have elevated levels of antibodies against antigens from Proteus mirabilis. Clin Rheumatol 2016; 36:527-535. [DOI: 10.1007/s10067-016-3441-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 09/26/2016] [Accepted: 09/28/2016] [Indexed: 10/20/2022]
|
29
|
Diamanti AP, Manuela Rosado M, Laganà B, D’Amelio R. Microbiota and chronic inflammatory arthritis: an interwoven link. J Transl Med 2016; 14:233. [PMID: 27492386 PMCID: PMC4973033 DOI: 10.1186/s12967-016-0989-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 07/26/2016] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Only recently, the scientific community gained insights on the importance of the intestinal resident flora for the host's health and disease. Gut microbiota in fact plays a crucial role in modulating innate and acquired immune responses and thus interferes with the fragile balance inflammation versus tolerance. MAIN BODY Correlations between gut bacteria composition and the severity of inflammation have been studied in inflammatory bowel diseases. More recently similar alterations in the gut microbiota have been reported in patients with spondyloarthritis, whereas in rheumatoid arthritis an accumulating body of evidence evokes a pathogenic role for the altered oral microbiota in disease development and course. In the context of dysbiosis it is also important to remember that different environmental factors like stress, smoke and dietary components can induce strong bacterial changes and consequent exposure of the intestinal epithelium to a variety of different metabolites, many of which have an unknown function. In this perspective, and in complex disorders like autoimmune diseases, not only the genetic makeup, sex and immunologic context of the individual but also the structure of his microbial community should be taken into account. CONCLUSIONS Here we provide a review of the role of the microbiota in the onset, severity and progression of chronic inflammatory arthritis as well as its impact on the therapeutic management of these patients. Furthermore we point-out the complex interwoven link between gut-joint-brain and immune system by reviewing the most recent data on the literature on the importance of environmental factors such as diet, smoke and stress.
Collapse
Affiliation(s)
- Andrea Picchianti Diamanti
- Department of Clinical and Molecular Sciences, II School of Medicine, S. Andrea University Hospital, “Sapienza” University of Rome, Rome, Italy
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, S. Andrea University Hospital, “Sapienza” University, Via di Grottarossa 1039, 00189 Rome, Italy
| | | | - Bruno Laganà
- Department of Clinical and Molecular Sciences, II School of Medicine, S. Andrea University Hospital, “Sapienza” University of Rome, Rome, Italy
| | - Raffaele D’Amelio
- Department of Clinical and Molecular Sciences, II School of Medicine, S. Andrea University Hospital, “Sapienza” University of Rome, Rome, Italy
| |
Collapse
|
30
|
Trela M, Nelson PN, Rylance PB. The role of molecular mimicry and other factors in the association of Human Endogenous Retroviruses and autoimmunity. APMIS 2016; 124:88-104. [PMID: 26818264 DOI: 10.1111/apm.12487] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 10/26/2015] [Indexed: 02/02/2023]
Abstract
Human Endogenous Retroviruses (HERVs) have been implicated in autoimmune and other diseases. Molecular mimicry has been postulated as a potential mechanism of autoimmunity. Exogenous viruses have also been reported to be associated with the same diseases, as have genetic and environmental factors. If molecular mimicry were to be shown to be an initiating mechanism of some autoimmune diseases, then therapeutic options of blocking antibodies and peptides might be of benefit in halting diseases at the outset. Bioinformatic and molecular modelling techniques have been employed to investigate molecular mimicry and the evidence for the association of HERVs and autoimmunity is reviewed. The most convincing evidence for molecular mimicry is in rheumatoid arthritis, where HERV K-10 shares amino acid sequences with IgG1Fc, a target for rheumatoid factor. Systemic lupus erythematosus is an example of a condition associated with several autoantibodies, and several endogenous and exogenous viruses have been reported to be associated with the disease. The lack of a clear link between one virus and this condition, and the spectrum of clinical manifestations, suggests that genetic, environmental and the inflammatory response to a virus or viruses might also be major factors in the pathogenesis of lupus and other autoimmune conditions. Where there are strong associations between a virus and an autoimmune condition, such as in hepatitis C and cryoglobulinaemia, the use of bioinformatics and molecular modelling can also be utilized to help to understand the role of molecular mimicry in how HERVs might trigger disease.
Collapse
Affiliation(s)
- Malgorzata Trela
- Immunology Research Group, Research Institute in Healthcare Sciences, University of Wolverhampton, Wolverhampton, UK
| | - Paul N Nelson
- Immunology Research Group, Research Institute in Healthcare Sciences, University of Wolverhampton, Wolverhampton, UK
| | - Paul B Rylance
- Royal Wolverhampton NHS Trust, New Cross Hospital, Wolverhampton, UK
| |
Collapse
|
31
|
The oral and gut microbiomes are perturbed in rheumatoid arthritis and partly normalized after treatment. Nat Med 2015. [PMID: 26214836 DOI: 10.1038/nm.3914] [Citation(s) in RCA: 1136] [Impact Index Per Article: 113.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We carried out metagenomic shotgun sequencing and a metagenome-wide association study (MGWAS) of fecal, dental and salivary samples from a cohort of individuals with rheumatoid arthritis (RA) and healthy controls. Concordance was observed between the gut and oral microbiomes, suggesting overlap in the abundance and function of species at different body sites. Dysbiosis was detected in the gut and oral microbiomes of RA patients, but it was partially resolved after RA treatment. Alterations in the gut, dental or saliva microbiome distinguished individuals with RA from healthy controls, were correlated with clinical measures and could be used to stratify individuals on the basis of their response to therapy. In particular, Haemophilus spp. were depleted in individuals with RA at all three sites and negatively correlated with levels of serum autoantibodies, whereas Lactobacillus salivarius was over-represented in individuals with RA at all three sites and was present in increased amounts in cases of very active RA. Functionally, the redox environment, transport and metabolism of iron, sulfur, zinc and arginine were altered in the microbiota of individuals with RA. Molecular mimicry of human antigens related to RA was also detectable. Our results establish specific alterations in the gut and oral microbiomes in individuals with RA and suggest potential ways of using microbiome composition for prognosis and diagnosis.
Collapse
|
32
|
Abstract
Deletion of the innate immune adaptor myeloid differentiation primary response gene 88 (MyD88) in the nonobese diabetic (NOD) mouse model of type 1 diabetes (T1D) results in microbiota-dependent protection from the disease: MyD88-negative mice in germ-free (GF), but not in specific pathogen-free conditions develop the disease. These results could be explained by expansion of particular protective bacteria ("specific lineage hypothesis") or by dominance of negative (tolerizing) signaling over proinflammatory signaling ("balanced signal hypothesis") in mutant mice. Here we found that colonization of GF mice with a variety of intestinal bacteria was capable of reducing T1D in MyD88-negative (but not wild-type NOD mice), favoring the balanced signal hypothesis. However, the receptors and signaling pathways involved in prevention or facilitation of the disease remained unknown. The protective signals triggered by the microbiota were revealed by testing NOD mice lacking MyD88 in combination with knockouts of several critical components of innate immune sensing for development of T1D. Only MyD88- and TIR-domain containing adapter inducing IFN β (TRIF) double deficient NOD mice developed the disease. Thus, TRIF signaling (likely downstream of Toll-like receptor 4, TLR4) serves as one of the microbiota-induced tolerizing pathways. At the same time another TLR (TLR2) provided prodiabetic signaling by controlling the microbiota, as reduction in T1D incidence caused by TLR2 deletion was reversed in GF TLR2-negative mice. Our results support the balanced signal hypothesis, in which microbes provide signals that both promote and inhibit autoimmunity by signaling through different receptors, including receptors of the TLR family.
Collapse
|
33
|
Slipping through the Cracks: Linking Low Immune Function and Intestinal Bacterial Imbalance to the Etiology of Rheumatoid Arthritis. Autoimmune Dis 2015; 2015:636207. [PMID: 25861466 PMCID: PMC4377354 DOI: 10.1155/2015/636207] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 12/05/2014] [Indexed: 12/22/2022] Open
Abstract
Autoimmune diseases (ADs) are considered to be caused by the host immune system which attacks and destroys its own tissue by mistake. A widely accepted hypothesis to explain the pathogenic mechanism of ADs is “molecular mimicry,” which states that antibodies against an infectious agent cross-react with a self-antigen sharing an identical or similar antigenic epitope. However, this hypothesis was most likely established based on misleading antibody assay data largely influenced by intense false positive reactions involved in immunoassay systems. Thus reinvestigation of this hypothesis using an appropriate blocking agent capable of eliminating all types of nonspecific reactions and proper assay design is strongly encouraged. In this review, we discuss the possibility that low immune function may be the fundamental, common defect in ADs, which increases the susceptibility to potential disease causative pathogens located in the gastrointestinal tract (GI), such as bacteria and their components or dietary components. In addition to these exogenous agents, aberrations in the host's physical condition may disrupt the host defense system, which is tightly orchestrated by “immune function,” “mucosal barrier function,” and “intestinal bacterial balance.” These disturbances may initiate a downward spiral, which can lead to chronic health problems that will evolve to an autoimmune disorder.
Collapse
|
34
|
Wang JJ, Yang GX, Zhang WC, Lu L, Tsuneyama K, Kronenberg M, Véla JL, Lopez-Hoyos M, He XS, Ridgway WM, Leung PSC, Gershwin ME. Escherichia coli infection induces autoimmune cholangitis and anti-mitochondrial antibodies in non-obese diabetic (NOD).B6 (Idd10/Idd18) mice. Clin Exp Immunol 2014; 175:192-201. [PMID: 24128311 DOI: 10.1111/cei.12224] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2013] [Indexed: 01/14/2023] Open
Abstract
Several epidemiological studies have demonstrated that patients with primary biliary cirrhosis (PBC) have a higher incidence of urinary tract infections (UTI) and there is significant homology of the immunodominant mitochondrial autoantigen, the E2 component of the pyruvate dehydrogenase complex (PDC-E2), between mammals and bacteria. Previous work has demonstrated that non-obese diabetic (NOD).B6 Idd10/Idd18 infected with Novosphingobium aromaticivorans developed liver lesions similar to human PBC. It was postulated that the biliary disease was dependent upon the presence of the unique N. aro glycosphingolipids in activating natural killer T (NK T) cells. To address this issue, we infected NOD.B6 Idd10/Idd18 mice with either Escherichia coli, N. aro or use of a phosphate-buffered saline (PBS) vehicle control and serially followed animals for the appearance of liver pathology and anti-mitochondrial autoantibodies (AMA). Of striking importance, the biliary disease of E. coli-infected mice was more severe than N. Aro-infected mice and the titre of AMA was higher in E. coli-infected mice. Furthermore, the immunopathology did not correlate with the ability of bacterial extracts to produce antigen-dependent activation of NK T cells. Our data suggest that the unique glycosphingolipids of N. aro are not required for the development of autoimmune cholangitis. Importantly, the data highlight the clinical significance of E. coli infection in a genetically susceptible host, and we suggest that the appearance of autoimmune cholangitis is dependent upon molecular mimicry. These data highlight that breach of tolerance to PDC-E2 is probably the first event in the natural history of PBC in genetically susceptible hosts.
Collapse
Affiliation(s)
- J J Wang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Tuuminen T, Lounamo K, Leirisalo-Repo M. A review of serological tests to assist diagnosis of reactive arthritis: critical appraisal on methodologies. Front Immunol 2013; 4:418. [PMID: 24363655 PMCID: PMC3849596 DOI: 10.3389/fimmu.2013.00418] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 11/16/2013] [Indexed: 01/17/2023] Open
Abstract
On a population-based level, the incidence of reactive arthritis (ReA) is 0.6–27/100,000. The definition of ReA varies and its pathogenesis is not yet clear. Attempts in basic immunology to suggest hypotheses for proliferation of forbidden B cell clones, molecular mimicry, and involvement of cross-reactive antibodies are helpful but not sufficient. Importantly, for the clinical diagnosis of the preceding infection, serology is widely used. Unfortunately, the accuracy of associations between serologic findings and clinical conclusions is plagued by poor standardization of methods. So far, few attempts have been done to examine the pitfalls of different approaches. Here, we review several serologic techniques, their performance and limitations. We will focus on serology for Yersinia, Campylobacter, Salmonella, Shigella, and Chlamydia trachomatis because these bacteria have a longer history of being associated with ReA. We also address controversies regarding the role of serology for some other bacteria linked to autoimmune disorders.
Collapse
Affiliation(s)
- Tamara Tuuminen
- Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki , Helsinki , Finland ; Eastern Finland Laboratory Centre Joint Authority Enterprise (ISLAB), Mikkeli District Laboratory , Mikkeli , Finland
| | - Kari Lounamo
- Department of Infectious Diseases, Health Centre of Lahti , Lahti , Finland
| | - Marjatta Leirisalo-Repo
- Institute of Clinical Medicine, University of Helsinki , Helsinki , Finland ; Department of Medicine, Helsinki University Central Hospital , Helsinki , Finland
| |
Collapse
|
36
|
Guarneri F, Guarneri C. Molecular mimicry in cutaneous autoimmune diseases. World J Dermatol 2013; 2:36-43. [DOI: 10.5314/wjd.v2.i4.36] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 09/17/2013] [Indexed: 02/06/2023] Open
Abstract
The emulation of characteristics of a different organism to gain biological advantage is a common phenomenon in nature, described and defined with the term “mimicry” in the second half of the 19th century. In the last decades, mimicry at molecular level has been evidenced as a method used by several pathogen microrganisms to control metabolic functions of infected cells and elude host’s immune system. Because of molecular mimicry, immune reactions against microbial molecules can turn against the mimicked self-molecules in predisposed subjects, leading to autoimmunity. This pathogenic mechanism, which gives a possible explanation for the specific epidemiological and chronological association between some infections and some autoimmune diseases, is well known and verified in many fields of medicine, but not adequately studied in dermatology: experimental data are available only for leprosy, atopic dermatitis, Behçet’s disease, Vogt-Koyanagi-Harada syndrome and systemic erythematous lupus, while for few other diseases its role is hypothetical or suggested on the basis of single, small experiments or anecdotal reports. An overview of available data and hypotheses about the role of molecular mimicry in autoimmune cutaneous diseases is presented here, together with the perspectives offered by the use of bioinformatics and the personal experience of the author in this field.
Collapse
|
37
|
Saba ES, Gueyffier L, Dichtel-Danjoy ML, Pozzetto B, Bourlet T, Gueyffier F, Mekki Y, Pottel H, Sabino EC, Vanhems P, Zrein MA. Anti-Trypanosoma cruzi cross-reactive antibodies detected at high rate in non-exposed individuals living in non-endemic regions: seroprevalence and association to other viral serologies. PLoS One 2013; 8:e74493. [PMID: 24069315 PMCID: PMC3775794 DOI: 10.1371/journal.pone.0074493] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 07/22/2013] [Indexed: 01/03/2023] Open
Abstract
Cross-reactive antibodies are characterized by their recognition of antigens that are different from the trigger immunogen. This happens when the similarity between two different antigenic determinants becomes adequate enough to enable a specific binding with such cross-reactive antibodies. In the present manuscript, we report the presence, at an “abnormal” high frequency, of antibodies in blood samples from French human subjects cross-reacting with a synthetic-peptide antigen derived from a Trypanosoma cruzi (T. cruzi) protein sequence. As the vector of T. cruzi is virtually confined to South America, the parasite is unlikely to be the trigger immunogen of the cross-reactive antibodies detected in France. At present, the cross-reactive antibodies are measured by using an in-house ELISA method that employs the T. cruzi -peptide antigen. However, to underline their cross-reactive characteristics, we called these antibodies “Trypanosoma cruzi Cross Reactive Antibodies” or TcCRA. To validate their cross-reactive nature, these antibodies were affinity-purified from plasma of healthy blood donor and were then shown to specifically react with the T. cruzi parasite by immunofluorescence. Seroprevalence of TcCRA was estimated at 45% in serum samples of French blood donors while the same peptide-antigen reacts with about 96% of T. cruzi -infected Brazilian individuals. In addition, we compared the serology of TcCRA to other serologies such as HSV 1/2, EBV, HHV-6, CMV, VZV, adenovirus, parvovirus B19, mumps virus, rubella virus, respiratory syncytial virus, measles and enterovirus. No association was identified to any of the tested viruses. Furthermore, we tested sera from different age groups for TcCRA and found a progressive acquisition starting from early childhood. Our findings show a large seroprevalence of cross-reactive antibodies to a well-defined T. cruzi antigen and suggest they are induced by a widely spread immunogen, acquired from childhood. The etiology of TcCRA and their clinical relevance still need to be investigated.
Collapse
Affiliation(s)
- Esber S. Saba
- InfYnity-Biomarkers, Ecully, France
- Laboratory of Bacteriology-Virology, GIMAP EA3064, Faculty of Medicine Jacques Lisfranc, Saint-Etienne, France
| | | | | | - Bruno Pozzetto
- Laboratory of Bacteriology-Virology, GIMAP EA3064, Faculty of Medicine Jacques Lisfranc, Saint-Etienne, France
| | - Thomas Bourlet
- Laboratory of Bacteriology-Virology, GIMAP EA3064, Faculty of Medicine Jacques Lisfranc, Saint-Etienne, France
| | - François Gueyffier
- UCBL-Hospices Civils de Lyon, Faculty of Medicine Rockefeller, Lyon, France
| | - Yahia Mekki
- UCBL-Hospices Civils de Lyon, Faculty of Medicine Rockefeller, Lyon, France
| | - Hans Pottel
- Interdisciplinary Research Center, Catholic University Leuven, Campus Kortrijk, Kortrijk, Belgium
| | - Ester C. Sabino
- Faculdade de Medicina da USP, Dep de Molestias Infecciosas e Parasitárias, São Paulo, Brazil
| | - Philippe Vanhems
- UCBL-Hospices Civils de Lyon, Faculty of Medicine Rockefeller, Lyon, France
| | - Maan A. Zrein
- InfYnity-Biomarkers, Ecully, France
- UCBL-Hospices Civils de Lyon, Faculty of Medicine Rockefeller, Lyon, France
- * E-mail:
| |
Collapse
|
38
|
Vaishnav RA, Liu R, Chapman J, Roberts AM, Ye H, Rebolledo-Mendez JD, Tabira T, Fitzpatrick AH, Achiron A, Running MP, Friedland RP. Aquaporin 4 molecular mimicry and implications for neuromyelitis optica. J Neuroimmunol 2013; 260:92-8. [PMID: 23664693 PMCID: PMC3682654 DOI: 10.1016/j.jneuroim.2013.04.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 04/10/2013] [Accepted: 04/11/2013] [Indexed: 12/31/2022]
Abstract
Neuromyelitis optica (NMO) is associated with antibodies to aquaporin 4 (AQP4). We hypothesized that antibodies to AQP4 can be triggered by exposure to environmental proteins. We compared human AQP4 to plant and bacterial proteins to investigate the occurrence of significantly similar structures and sequences. High similarity to a known epitope for NMO-IgG, AQP4(207-232), was observed for corn ZmTIP4-1. NMO and non-NMO sera were assessed for reactivity to AQP4(207-232) and the corn peptide. NMO patient serum showed reactivity to both peptides as well as to plant tissue. These findings warrant further investigation into the role of the environment in NMO etiology.
Collapse
Affiliation(s)
- Radhika A. Vaishnav
- Department of Neurology, University of Louisville, KY, USA
- Department of Physiology and Biophysics, University of Louisville, KY, USA
| | - Ruolan Liu
- Department of Neurology, University of Louisville, KY, USA
| | - Joab Chapman
- Department of Neurology, Sheba Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Israel
| | - Andrew M. Roberts
- Department of Physiology and Biophysics, University of Louisville, KY, USA
| | - Hong Ye
- Department of Pharmacology, University of Louisville, KY, USA
| | | | - Takeshi Tabira
- Department of Diagnosis, Prevention, and Treatment of Dementia, Graduate School of Juntendo University, Tokyo, Japan
| | | | - Anat Achiron
- Department of Neurology, Sheba Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Israel
| | | | - Robert P. Friedland
- Department of Neurology, University of Louisville, KY, USA
- Department of Biochemistry, University of Louisville, KY, USA
| |
Collapse
|
39
|
Schaeverbeke T, Truchetet ME, Richez C. Gut metagenome and spondyloarthritis. Joint Bone Spine 2013; 80:349-52. [PMID: 23806346 DOI: 10.1016/j.jbspin.2013.02.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2013] [Indexed: 02/07/2023]
|
40
|
Rashid T, Wilson C, Ebringer A. The link between ankylosing spondylitis, Crohn's disease, Klebsiella, and starch consumption. Clin Dev Immunol 2013; 2013:872632. [PMID: 23781254 PMCID: PMC3678459 DOI: 10.1155/2013/872632] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Accepted: 04/23/2013] [Indexed: 12/22/2022]
Abstract
Both ankylosing spondylitis (AS) and Crohn's disease (CD) are chronic and potentially disabling interrelated conditions, which have been included under the group of spondyloarthropathies. The results of a large number of studies support the idea that an enteropathic pathogen, Klebsiella pneumoniae, is the most likely triggering factor involved in the initiation and development of these diseases. Increased starch consumptions by genetically susceptible individuals such as those possessing HLA-B27 allelotypes could trigger the disease in both AS and CD by enhancing the growth and perpetuation of the Klebsiella microbes in the bowel. Exposure to increased levels of these microbes will lead to the production of elevated levels of anti-Klebsiella antibodies as well as autoantibodies against cross-reactive self-antigens with resultant pathological lesions in the bowel and joints. Hence, a decrease of starch-containing products in the daily dietary intake could have a beneficial therapeutic effect on the disease especially when used in conjunction with the currently available medical therapies in the treatment of patients with AS and CD.
Collapse
Affiliation(s)
- Taha Rashid
- Analytical Sciences Group, Kings College, 150 Stamford Street, London SE1 9NH, UK
| | - Clyde Wilson
- Department of Pathology and Microbiology, Kings Edward VII Memorial Hospital, 7 Point Finger Road, Paget DV04, Bermuda
| | - Alan Ebringer
- Analytical Sciences Group, Kings College, 150 Stamford Street, London SE1 9NH, UK
| |
Collapse
|
41
|
Abstract
The commensal microbiota affects many aspects of mammalian health including control of the immune system to such a extent that a "commensalocentric" view of the maintenance of overall health could be suggested. Autoimmunity is a case of mistaken identity: The immune system reacts to self-tissues and cells as if they were pathogens. Autoimmune reactions can be both advanced or blocked by the commensal microbiota, which can affect innate and adaptive arms of immune responses as well as the mechanisms of "innate-adaptive connection." Whether specific microbial lineages affect immunity and autoimmunity (the "specific lineage hypothesis") or multiple lineages can tip the homeostatic balance that regulates host/microbiota homeostasis toward reduced or enhanced host reactivity (the "balanced signal hypothesis") is yet unknown. The complexity of host/microbiota interactions needs to be fully appreciated in order to find the means for prophylaxis and treatment of autoimmune disorders.
Collapse
|
42
|
Kean WF, Tocchio S, Kean M, Rainsford KD. The musculoskeletal abnormalities of the Similaun Iceman ("ÖTZI"): clues to chronic pain and possible treatments. Inflammopharmacology 2013; 21:11-20. [PMID: 23096483 PMCID: PMC3560943 DOI: 10.1007/s10787-012-0153-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 09/21/2012] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND INTRODUCTION In 1991, a deceased human male was found frozen in a glacier pool in the Italian Alps in north west Italy, and is now carefully preserved in the South Tyrol Museum of Archaeology, in Bolzano, Italy. The bodily tissues of the 5,300 year old male (colloquially referred to as the Iceman or Ötzi) were well preserved despite damage related to freezing, and glacial movement. Associated articles of well-preserved clothing, tools, weapons and other devices were also present and have been studied in detail. Clinical examination and imaging investigations have also shown that the Icemen had experienced possible illnesses in his lifetime and had identifiable areas of arthritis and musculoskeletal injury. This report includes some key observations on the musculoskeletal state of Ötzi and reference to the involvement of tattoo markings. Some aspects about the aetiology of his abnormalities and inflammatory arthritis are considered along with possible treatments that he might have employed. METHODS AND RESULTS We (WFK and MK) undertook a clinical musculoskeletal examination of the Iceman, details of which with available photographs and radiographic imaging pertaining to the musculoskeletal findings of the Iceman are reported here. The skin of the Iceman has numerous linear carbon tattoos, which are not of a decorative type. These have been presumed to possibly be "medicinal" tattoos administered for therapeutic reasons and may have been used in acupuncture-like treatment of pain. Spinal imaging identified areas of spinal damage and our observations have provided clues as to possible sites of spinal initiated pain and hence sites for administration of the "medicinal" tattoos. We observed body areas of the Iceman, in which imaging demonstrated arthritis and other forms of long-term musculoskeletal damage, but which do not have adjacent or corresponding "medicinal" tattoos. We contend that the back and leg "medicinal" tattoos correspond directly to sites of chronic right knee and right ankle pain, and left thoracolumbar pain. They also correspond to lower lumbar and sciatic referred radicular pain which may have a contributory cause related to the presence of a transitional lumbar 5 vertebra. Using recent published data (Keller et al. in Nature Commun 3:698, 2012. doi: 10.1038/ncomms1701 ) of the genome structure of the Iceman, we suggest some potential causes of the osteoarthritis or inflammatory joint injury may relate to presence of coronary heart disease (CHD) and Lyme disease (Borrelia burgdorferi) infection. We speculate on possible medical applications of natural products for self-medication. CONCLUSIONS These observations highlight several diagnostic features of musculoskeletal conditions in the Iceman with the possibility that tattoos may have been used for diagnosis or location of his painful states. The origins of his musculoskeletal conditions are unclear but there are indications that Lyme disease and CHD may have been factors. The associations or use of natural products may give insights into their applications at the time of the life of the Iceman.
Collapse
Affiliation(s)
- Walter F Kean
- Division of Rheumatology, McMaster University, 401-1 Young Street, Hamilton, ON, L8N1T8, Canada.
| | | | | | | |
Collapse
|
43
|
Environment and primary biliary cirrhosis: electrophilic drugs and the induction of AMA. J Autoimmun 2013; 41:79-86. [PMID: 23352659 DOI: 10.1016/j.jaut.2012.12.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 12/17/2012] [Accepted: 12/20/2012] [Indexed: 12/12/2022]
Abstract
Environmental stimulation is a major factor in the initiation and perpetuation of autoimmune diseases. We have addressed this issue and focused on primary biliary cirrhosis (PBC), an autoimmune disease of the liver. Immunologically, PBC is distinguished by immune mediated destruction of the intra hepatic bile ducts and the presence of high titer antimitochondrial autoantibodies (AMA) directed against a highly specific epitope within the lipoic acid binding domain of the pyruvate dehydrogenase E2 subunit (PDC-E2). We submit that the uniqueness of AMA epitope specificity and the conformational changes of the PDC-E2 lipoyl domain during physiological acyl transfer could be the lynchpin to the etiology of PBC and postulate that chemical xenobiotics modification of the lipoyl domain of PDC-E2 is sufficient to break self-tolerance, with subsequent production of AMA in patients with PBC. Indeed, using quantitative structure activity relationship (QSAR) analysis on a peptide-xenobiotic conjugate microarray platform, we have demonstrated that when the lipoyl domain of PDC-E2 was modified with specific synthetic small molecule lipoyl mimics, the ensuing structures displayed highly specific reactivity to PBC sera, at levels often higher than the native PDC-E2 molecule. Hereby, we discuss our recent QSAR analysis data on specific AMA reactivity against a focused panel of lipoic acid mimic in which the lipoyl di-sulfide bond are modified. Furthermore, data on the immunological characterization of antigen and Ig isotype specificities against one such lipoic acid mimic; 6,8-bis(acetylthio)octanoic acid (SAc), when compared with rPDC-E2, strongly support a xenobiotic etiology in PBC. This observation is of particular significance in that approximately one third of patients who have taken excessive acetaminophen (APAP) developed AMA with same specificity as patients with PBC, suggesting that the lipoic domain are a target of APAP electrophilic metabolites such as NAPQI. We submit that in genetically susceptible hosts, electrophilic modification of lipoic acid in PDC-E2 by acetaminophen or similar drugs can facilitate loss of tolerance and lead to the development of PBC.
Collapse
|
44
|
Distinct mechanisms mediate naive and memory CD8 T-cell tolerance. Proc Natl Acad Sci U S A 2012; 109:21438-43. [PMID: 23236165 DOI: 10.1073/pnas.1217409110] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Peripheral tolerance to developmentally regulated antigens is necessary to sustain tissue homeostasis. We have now devised an inducible and reversible system that allows interrogation of T-cell tolerance induction in endogenous naïve and memory CD8 T cells. Our data show that peripheral CD8 T-cell tolerance can be preserved through two distinct mechanisms, antigen addiction leading to anergy for naïve T cells and ignorance for memory T cells. Induction of antigen in dendritic cells resulted in substantial expansion and maintenance of endogenous antigen-specific CD8 T cells. The self-reactive cells initially exhibited effector activity but eventually became unresponsive. Upon antigen removal, the antigen-specific population waned, resulting in development of a self-specific memory subset that recalled to subsequent challenge. In striking contrast to naïve CD8 T cells, preexisting antigen-specific memory CD8 T cells failed to expand after antigen induction and essentially ignored the antigen despite widespread expression by dendritic cells. The inclusion of inflammatory signals partially overcame memory CD8 T-cell ignorance of self-antigen. Thus, peripheral CD8 T-cell tolerance for naïve CD8 T cells depended on the continuous presence of antigen, whereas memory CD8 T cells were prohibited from autoreactivity in the absence of inflammation.
Collapse
|
45
|
H11/HspB8 and Its Herpes Simplex Virus Type 2 Homologue ICP10PK Share Functions That Regulate Cell Life/Death Decisions and Human Disease. Autoimmune Dis 2012; 2012:395329. [PMID: 23056924 PMCID: PMC3463903 DOI: 10.1155/2012/395329] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 08/20/2012] [Accepted: 08/21/2012] [Indexed: 12/24/2022] Open
Abstract
Small heat shock proteins (sHsp) also known as HspB are a large family of widely expressed proteins that contain a 90 residues domain known as α-crystallin. Here, we focus on the family member H11/HspB8 and its herpes simplex virus type 2 (HSV-2) homologue ICP10PK, and discuss the possible impact of this relationship on human disease. H11/HspB8 and ICP10PK are atypical protein kinases. They share multi-functional activity that encompasses signaling, unfolded protein response (UPR) and the regulation of life cycle potential. In melanocytes H11/HspB8 causes growth arrest. It is silenced in a high proportion of melanoma prostate cancer, Ewing's sarcoma and hematologic malignancies through aberrant DNA methylation. Its restored expression induces cell death and inhibits tumor growth in xenograft models, identifying H11/HspB8 as a tumor suppressor. This function involves the activation of multiple and distinct death pathways, all of which initiate with H11/HspB8-mediated phosphorylation of transforming growth factor β-activated kinase 1 (TAK1). Both ICP10PK and H11/HspB8 were implicated in inflammatory processes that involve dendritic cells activation through Toll-like receptor-dependent pathways and may contribute to the onset of autoimmunity. The potential evolutionary relationship of H11/HspB8 to ICP10PK, its impact on human disorders and the development of therapeutic strategies are discussed.
Collapse
|
46
|
Bax HJ, Keeble AH, Gould HJ. Cytokinergic IgE Action in Mast Cell Activation. Front Immunol 2012; 3:229. [PMID: 22888332 PMCID: PMC3412263 DOI: 10.3389/fimmu.2012.00229] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 07/14/2012] [Indexed: 12/20/2022] Open
Abstract
Some 10 years ago it emerged that at sufficiently high concentrations certain monoclonal mouse IgEs exert previously unsuspected effects on mast cells. Thus they can both promote survival and induce activation of mast cells without the requirement for antigens. This was a wake up call that appears to have been missed (or dismissed) by the majority of immunologists. The structural attributes responsible for the potency of the so-called “highly cytokinergic” or HC IgEs have not yet been determined, but the events that ensue when such IgEs bind to the high-affinity receptor, FcεRI, on mast cells have been thoroughly studied, and are strikingly similar to those engendered by antigens when they form cross-linked complexes with the receptors. We review the evidence for the cytokinergic activity of IgE, and the structural features and known properties of immunoglobulins, and of IgE in particular, most likely to be implicated in the phenomenon. We suggest that IgEs with cytokinergic activity may be generated by local germinal center reactions in the target organs of allergy. We consider also the important implications that the existence of cytokinergic IgE may have for a fuller understanding of adaptive immunity and of the action of IgE in asthma and other diseases.
Collapse
Affiliation(s)
- Heather J Bax
- Randall Division of Cell and Molecular Biophysics, King's College London London, UK
| | | | | |
Collapse
|
47
|
Abstract
Most infectious agents, such as viruses, bacteria and parasites, can trigger autoimmunity via different mechanisms. The development of an autoimmune disorder after infection tends to occur in genetically susceptible individuals. Some parameters, such as genetic predisposition, feature of the infectious agent and sometimes protective effect of the infections, have a significant role in this process. These parameters and various pathogens that could lead to enhancement or exacerbation of autoimmune disease were examined in this review. Recent studies were reviewed from a microbiological perspective.
Collapse
|