1
|
Liu B, Wang K, Yang L, Zheng J, Ma T, Zhang S, Huang L, Chen T, Guo Y, Cui Z, Zhang X, Chen J, Lu H. 6-formylindolo[3, 2-b]carbazole alters gut microbiota and prevents the progression of ankylosing spondylitis in mice. Int Immunopharmacol 2024; 128:111562. [PMID: 38244515 DOI: 10.1016/j.intimp.2024.111562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 01/22/2024]
Abstract
Ankylosing spondylitis (AS), is known as a chronic inflammatory autoimmune disease, there is evidence to suggest that gut microbiota disorders may be related to the occurrence and development of AS. Studies have shown that 6-formylindolo[3, 2-b]carbazole (FICZ) has the ability to modulate intestinal homeostasis and inhibit inflammatory responses. The purpose of this work is to evaluate the protective role of FICZ in treating AS and elucidate potential mechanisms. FICZ was administered to the proteoglycan (PG)-induced AS mice for 7 consecutive weeks. The effects of FICZ on AS mice were evaluated by the disease severity, intestinal histopathology, proinflammatory cytokine levels, and intestinal mucosal barrier function. The gut microbiota compositions were profiled through 16S rDNA high-throughput sequencing. We found that FICZ significantly reduced the severity of AS and resulted in the downregulating of TNF-α and IL-17A inflammatory cytokines. Moreover, FICZ ameliorated pathological changes in the ileal and improved intestinal mucosal barrier function. Furthermore, FICZ altered the composition of the gut microbiota by increasing the Bacteroidetes/Firmicutes phylum ratio and enriched the genes related to "glycan biosynthesis and metabolism", thus reversing the process of AS. In conclusion, FICZ suppressed the progression of AS and altered gut microbiota in AS mice, which provided new insight into AS therapy strategy.
Collapse
Affiliation(s)
- Bin Liu
- Department of Spine Surgery, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Kun Wang
- Department of Spine Surgery, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China.
| | - Lianjun Yang
- Department of Spine Surgery, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Junchi Zheng
- Department of Orthopedics, Zhongshan Torch Development Zone People's Hospital, Zhongshan, Guangdong 528437, China; Department of Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510630, China
| | - Tao Ma
- Department of Biobank, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Shiyanjin Zhang
- Department of Spine Surgery, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Lihua Huang
- Department of Spine Surgery, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Tao Chen
- Department of Spine Surgery, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Yuanqing Guo
- Department of Spine Surgery, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Zhifei Cui
- Department of Spine Surgery, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Xueling Zhang
- Department of Child Healthcare, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Junquan Chen
- Department of Spine Surgery, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China.
| | - Hai Lu
- Department of Spine Surgery, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China.
| |
Collapse
|
2
|
Alfuzaie R. The Link Between Gastrointestinal Microbiome and Ocular Disorders. Clin Ophthalmol 2023; 17:2133-2140. [PMID: 37521153 PMCID: PMC10386868 DOI: 10.2147/opth.s415425] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 07/18/2023] [Indexed: 08/01/2023] Open
Abstract
The gut-eye axis has been hypothesized to be a factor in many eye pathologies. This review examines papers from PubMed about this topic. Bacterial commensals could either be protective by regulating the immune system or prove to be damaging to the gut mucosal wall and incite an inflammatory process. The balance between the two appears to be crucial in maintaining eye health. Imbalances have been implicated in ophthalmologic conditions. The use of probiotics, dietary modifications, antibiotics, and faecal microbiota transplant in mice with pathologies such as those encountered in our practice appears to reverse disease course or at least prevent its progression. Clinical trials are currently underway to investigate their clinical significance in diseased patients.
Collapse
|
3
|
Gill T, Stauffer P, Asquith M, Laderas T, Martin TM, Davin S, Schleisman M, Ramirez C, Ogle K, Lindquist I, Nguyen J, Planck SR, Shaut C, Diamond S, Rosenbaum JT, Karstens L. Axial spondyloarthritis patients have altered mucosal IgA response to oral and fecal microbiota. Front Immunol 2022; 13:965634. [PMID: 36248884 PMCID: PMC9556278 DOI: 10.3389/fimmu.2022.965634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/19/2022] [Indexed: 11/23/2022] Open
Abstract
Axial spondyloarthritis (axSpA) is an inflammatory arthritis involving the spine and the sacroiliac joint with extra-articular manifestations in the eye, gut, and skin. The intestinal microbiota has been implicated as a central environmental component in the pathogenesis of various types of spondyloarthritis including axSpA. Additionally, alterations in the oral microbiota have been shown in various rheumatological conditions, such as rheumatoid arthritis (RA). Therefore, the aim of this study was to investigate whether axSpA patients have an altered immunoglobulin A (IgA) response in the gut and oral microbial communities. We performed 16S rRNA gene (16S) sequencing on IgA positive (IgA+) and IgA negative (IgA-) fractions (IgA-SEQ) from feces (n=17 axSpA; n=14 healthy) and saliva (n=14 axSpA; n=12 healthy), as well as on IgA-unsorted fecal and salivary samples. PICRUSt2 was used to predict microbial metabolic potential in axSpA patients and healthy controls (HCs). IgA-SEQ analyses revealed enrichment of several microbes in the fecal (Akkermansia, Ruminococcaceae, Lachnospira) and salivary (Prevotellaceae, Actinobacillus) microbiome in axSpA patients as compared with HCs. Fecal microbiome from axSpA patients showed a tendency towards increased alpha diversity in IgA+ fraction and decreased diversity in IgA- fraction in comparison with HCs, while the salivary microbiome exhibits a significant decrease in alpha diversity in both IgA+ and IgA- fractions. Increased IgA coating of Clostridiales Family XIII in feces correlated with disease severity. Inferred metagenomic analysis suggests perturbation of metabolites and metabolic pathways for inflammation (oxidative stress, amino acid degradation) and metabolism (propanoate and butanoate) in axSpA patients. Analyses of fecal and salivary microbes from axSpA patients reveal distinct populations of immunoreactive microbes compared to HCs using the IgA-SEQ approach. These bacteria were not identified by comparing their relative abundance alone. Predictive metagenomic analysis revealed perturbation of metabolites/metabolic pathways in axSpA patients. Future studies on these immunoreactive microbes may lead to better understanding of the functional role of IgA in maintaining microbial structure and human health.
Collapse
Affiliation(s)
- Tejpal Gill
- Division of Arthritis and Rheumatic Diseases, Department of Medicine, Oregon Health & Science University, Portland, OR, United States
- *Correspondence: Tejpal Gill,
| | - Patrick Stauffer
- Casey Eye Institute/Department of Ophthalmology, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Mark Asquith
- Division of Arthritis and Rheumatic Diseases, Department of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Ted Laderas
- Division of Bioinformatics and Computational Biomedicine, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, United States
| | - Tammy M. Martin
- Casey Eye Institute/Department of Ophthalmology, School of Medicine, Oregon Health & Science University, Portland, OR, United States
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, United States
| | - Sean Davin
- Casey Eye Institute/Department of Ophthalmology, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Matthew Schleisman
- Casey Eye Institute/Department of Ophthalmology, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Claire Ramirez
- Casey Eye Institute/Department of Ophthalmology, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Kimberly Ogle
- Casey Eye Institute/Department of Ophthalmology, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Ingrid Lindquist
- Division of Arthritis and Rheumatic Diseases, Department of Medicine, Oregon Health & Science University, Portland, OR, United States
- Department of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Justine Nguyen
- Division of Bioinformatics and Computational Biomedicine, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, United States
| | - Stephen R. Planck
- Casey Eye Institute/Department of Ophthalmology, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Carley Shaut
- Laboratory of Immunogenetics, Oregon Health & Science University, Portland, OR, United States
| | - Sarah Diamond
- Department of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - James T. Rosenbaum
- Division of Arthritis and Rheumatic Diseases, Department of Medicine, Oregon Health & Science University, Portland, OR, United States
- Casey Eye Institute/Department of Ophthalmology, School of Medicine, Oregon Health & Science University, Portland, OR, United States
- Department of Cell Biology, Oregon Health & Science University, Portland, OR, United States
- Legacy Devers Eye Institute, Portland, OR, United States
| | - Lisa Karstens
- Division of Bioinformatics and Computational Biomedicine, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, United States
- Division of Urogynecology, Department of Obstetrics and Gynecology Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
4
|
Kalogeropoulos D, Barry R, Kalogeropoulos C. The association between intestinal microbiome and autoimmune uveitis. ARCHIVOS DE LA SOCIEDAD ESPANOLA DE OFTALMOLOGIA 2022; 97:264-275. [PMID: 35526950 DOI: 10.1016/j.oftale.2021.01.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 01/21/2021] [Indexed: 06/14/2023]
Abstract
INTRODUCTION AND OBJECTIVES The microbiome is strongly implicated in a wide spectrum of immune-mediated diseases, whereas gut commensal microbiota plays a pivotal role in immune and intestinal homeostasis. MATERIALS AND METHODS A thorough literature search was performed in PubMed database. An additional search was made in Google Scholar to complete the collected items. RESULTS Due to complex interactions with the host genetics and other factors, intestinal dysbiosis has been linked to various immune-mediated disorders. In particular, the role of intestinal microbiota in the pathogenesis of uveitis has been demonstrated by several studies, indicating that changes in the microbiome can trigger autoimmune ocular inflammatory processes or affect their severity. CONCLUSIONS This review summarizes how alterations in the intestinal microbiota can conduce to immune-mediated ocular pathologies and how microbiome can be targeted in order to form novel therapeutic approaches to treat these severe and potentially blinding conditions.
Collapse
Affiliation(s)
- D Kalogeropoulos
- Department of Ophthalmology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece.
| | - R Barry
- Institute of Clinical Sciences, University of Birmingham, College of Medical and Dental Sciences, Birmingham, United Kingdom; Department of Ophthalmology, Birmingham & Midland Eye Centre, Sandwell & West Birmingham Hospitals NHS Trust, Birmingham, United Kingdom
| | - C Kalogeropoulos
- Department of Ophthalmology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| |
Collapse
|
5
|
Sagard J, Olofsson T, Mogard E, Marsal J, Andréasson K, Geijer M, Kristensen LE, Lindqvist E, Wallman JK. Gut dysbiosis associated with worse disease activity and physical function in axial spondyloarthritis. Arthritis Res Ther 2022; 24:42. [PMID: 35151357 PMCID: PMC8840679 DOI: 10.1186/s13075-022-02733-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/31/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Based on clinical and genetic associations, axial spondyloarthritis (axSpA) and inflammatory bowel disease (IBD) are suspected to have a linked pathogenesis. Gut dysbiosis, intrinsic to IBD, has also been observed in axSpA. It is, however, not established to what degree gut dysbiosis is associated with axSpA disease severity. The objective of this study was to compare gut dysbiosis frequency between controls, non-radiographic axial spondyloarthritis (nr-axSpA), and ankylosing spondylitis (AS) patients and investigate whether gut dysbiosis is cross-sectionally associated with axSpA disease activity, physical function, mobility, or pain. METHODS Gut dysbiosis was assessed by 16SrRNA analysis of feces from 44/88 nr-axSpA/AS patients (ASAS/mNY criteria) without inflammatory bowel disease (IBD) and 46 controls without IBD or rheumatic disease. The GA-map™ Dysbiosis Test was used, grading gut microbiota aberrations on a 1-5 scale, where ≥3 denotes dysbiosis. Proportions with dysbiosis were compared between the groups. Furthermore, standard axSpA measures of disease activity, function, mobility, and pain were compared between patients (nr-axSpA and AS combined) with and without dysbiosis, univariately, and adjusted for relevant confounders (ANCOVA). RESULTS Gut dysbiosis was more frequent in AS than controls (36% versus 17%, p=0.023), while nr-axSpA (25% dysbiosis) did not differ significantly from either AS or controls. Univariately, most axSpA measures were significantly worse in patients with dysbiosis versus those without: ASDAS-CRP between-group difference 0.6 (95% CI 0.2-0.9); BASDAI 1.6 (0.8-2.4); evaluator's global disease activity assessment (Likert scale 0-4) 0.3 (0.1-0.5), BASFI 1.5 (0.6-2.4), and VAS pain (cm) 1.3 (0.4-2.2). Differences remained significant after adjustment for demographics, lifestyle factors, treatments, gut inflammation (fecal calprotectin ≥50 mg/kg), and gut symptoms, except for VAS pain. BASMI and CRP were not associated with dysbiosis. CONCLUSION Gut dysbiosis, more frequent in AS patients than controls, is associated with worse axSpA disease activity and physical function, seemingly irrespective of both gut inflammation and treatments. This provides further evidence for an important link between disturbances in gastrointestinal homeostasis and axSpA.
Collapse
Affiliation(s)
- Jonas Sagard
- Section of Rheumatology, Department of Clinical Sciences Lund, Lund University, Kioskgatan 5, 22185, Lund, Sweden.
- Skåne University Hospital, Department of Rheumatology, Lund, Sweden.
| | - Tor Olofsson
- Section of Rheumatology, Department of Clinical Sciences Lund, Lund University, Kioskgatan 5, 22185, Lund, Sweden
- Skåne University Hospital, Department of Rheumatology, Lund, Sweden
| | - Elisabeth Mogard
- Section of Rheumatology, Department of Clinical Sciences Lund, Lund University, Kioskgatan 5, 22185, Lund, Sweden
- Skåne University Hospital, Department of Rheumatology, Lund, Sweden
| | - Jan Marsal
- Department of Immunology, EMV, Lund University, Lund, Sweden
- Department of Gastroenterology, Skåne University Hospital, Lund/Malmö, Sweden
| | - Kristofer Andréasson
- Section of Rheumatology, Department of Clinical Sciences Lund, Lund University, Kioskgatan 5, 22185, Lund, Sweden
- Skåne University Hospital, Department of Rheumatology, Lund, Sweden
| | - Mats Geijer
- Sahlgrenska Academy, Institute of Clinical Sciences, Department of Radiology, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Department of Radiology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Section of Radiology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Lars Erik Kristensen
- Section of Rheumatology, Department of Clinical Sciences Lund, Lund University, Kioskgatan 5, 22185, Lund, Sweden
- Parker Institute, Frederiksberg and Bispebjerg, Department of Rheumatology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Elisabet Lindqvist
- Section of Rheumatology, Department of Clinical Sciences Lund, Lund University, Kioskgatan 5, 22185, Lund, Sweden
- Skåne University Hospital, Department of Rheumatology, Lund, Sweden
| | - Johan K Wallman
- Section of Rheumatology, Department of Clinical Sciences Lund, Lund University, Kioskgatan 5, 22185, Lund, Sweden
- Skåne University Hospital, Department of Rheumatology, Lund, Sweden
| |
Collapse
|
6
|
Zhang X, Sun Z, Zhou A, Tao L, Chen Y, Shi X, Yin J, Sun Z, Ding G. Association Between Infections and Risk of Ankylosing Spondylitis: A Systematic Review and Meta-Analysis. Front Immunol 2021; 12:768741. [PMID: 34745144 PMCID: PMC8569302 DOI: 10.3389/fimmu.2021.768741] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/11/2021] [Indexed: 12/17/2022] Open
Abstract
Background Previous literature on the association between infections and the risk of developing ankylosing spondylitis (AS) presented controversial results. This meta-analysis aimed to quantitatively investigate the effect of infections on the risk of AS. Methods We searched the PubMed, Embase, and Web of Science databases until March 26, 2021 for analytical epidemiological studies on the association between infections and the risk of AS. Fixed or random effect models were used to calculate total risk estimates based on study heterogeneity. Subgroup analysis, and sensitivity analysis were also performed. Publication bias was estimated using funnel plots and Begg’s test. Results Six case-control articles (n=1,296,239) and seven cohort articles (n=7,618,524) were incorporated into our meta-analysis. The pooled odds ratio (OR) from these case-control studies showed that infections were associated with an increased risk of AS (OR=1.46, 95% confidence interval [CI], 1.23–1.73), and the pooled relative risk (RR) from the cohort studies showed the same findings (RR=1.35, 95% CI, 1.12–1.63). Subgroup analysis showed that infections in participants with unadjusted comorbidities (OR=1.66, 95% CI, 1.35–2.03), other types of infection (OR=1.40, 95% CI, 1.15–1.70), and infection of the immune system (OR=1.46, 95% CI, 1.42–1.49) were associated with the risk of AS in case-control studies. In cohort studies, infections with adjusted comorbidities (RR=1.39, 95% CI, 1.15–1.68), viral infection (RR=1.43, 95% CI, 1.22–1.66), other types of infection (RR=1.44, 95% CI, 1.12–1.86), and other sites of infection (RR=1.36, 95% CI, 1.11–1.67) were associated with an increased risk of AS. Conclusions The findings of this meta-analysis confirm that infections significantly increase the risks of AS. This is helpful in providing an essential basis for the prevention of AS via the avoidance of infections.
Collapse
Affiliation(s)
- Xiao Zhang
- School of Nursing, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China.,School of Clinical Medline, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.,School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Zhe Sun
- The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Aihong Zhou
- The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Lei Tao
- The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Yingxin Chen
- School of Nursing, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China.,School of Clinical Medline, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xinyu Shi
- School of Nursing, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China.,School of Clinical Medline, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Jia Yin
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Zheng Sun
- School of Clinical Medline, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Guoyong Ding
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China.,The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| |
Collapse
|
7
|
Abstract
Reactive arthritis (ReA) following bacterial infection from the urogenital and gastrointestinal tract is widely described but is not typical post-viral infections. This report presents the second case of ReA after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in the United States. A 45-year-old black male with chronic low back pain was hospitalized for 45 days with coronavirus disease 2019 (COVID-19), complicated due to the development of multiorgan failure managed with intubation, extracorporeal membrane oxygenation, and hemodialysis. He was subsequently discharged to an acute rehabilitation facility where he complained of new-onset pain in his shoulders, left elbow, and left knee three weeks after a negative SARS-CoV-2 test. He was readmitted from his acute rehabilitation facility due to recurrent fever and the development of a swollen, warm left knee. Laboratory studies at readmission showed elevated inflammatory markers, negative extensive infectious disease workup, and aseptic inflammatory left knee synovial fluid without crystals. Testing returned negative for most common antibodies seen in immune-mediated arthritides (e.g., rheumatoid arthritis, systemic lupus erythematosus), as well as for common respiratory and gastrointestinal tract pathogens responsible for viral arthritis. The multidisciplinary inpatient medical team deemed the clinical presentation and laboratory findings most consistent with ReA. The patient received a course of oral corticosteroids, followed by a second course due to the recurrence of symptoms weeks after initial treatment and recovery. The current body of medical literature on SARS-CoV-2 pathophysiology supports plausible mechanisms on how this infection may induce ReA. Such a scenario should be considered in the differential of COVID-19-recovered patients presenting with polyarthritis as prompt steroid treatment may help patient recovery.
Collapse
Affiliation(s)
- Faizal Ouedraogo
- Department of Medicine, University of Maryland Capital Region Health, Largo, USA
| | - Rachita Navara
- Division of Cardiology, Washington University School of Medicine, St. Louis, USA
| | - Rusha Thapa
- Medicine, Nepal Medical School, Kathmandu City, NPL
| | - Kunj G Patel
- Department of Physical Medicine and Rehabilitation, Barnes-Jewish West County Hospital, St. Louis, USA
| |
Collapse
|
8
|
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
|
9
|
Papagoras C, Voulgari PV, Drosos AA. Cardiovascular Disease in Spondyloarthritides. Curr Vasc Pharmacol 2020; 18:473-487. [PMID: 31330576 DOI: 10.2174/1570161117666190426164306] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/13/2019] [Accepted: 04/13/2019] [Indexed: 12/15/2022]
Abstract
The spondyloarthritides are a group of chronic systemic inflammatory joint diseases, the main types being ankylosing spondylitis (AS) and psoriatic arthritis (PsA). Evidence accumulating during the last decades suggests that patients with AS or PsA carry an increased risk for cardiovascular disease and cardiovascular death. This risk appears to be mediated by systemic inflammation over and above classical cardiovascular risk factors. The excess cardiovascular risk in those patients has been formally acknowledged by scientific organizations, which have called physicians' attention to the matter. The application by Rheumatologists of new effective anti-rheumatic treatments and treat-to-target strategies seems to benefit patients from a cardiovascular point of view, as well. However, more data are needed in order to verify whether anti-rheumatic treatments do have an effect on cardiovascular risk and whether there are differences among them in this regard. Most importantly, a higher level of awareness of the cardiovascular risk is needed among patients and healthcare providers, better tools to recognize at-risk patients and, ultimately, commitment to address in parallel both the musculoskeletal and the cardiovascular aspect of the disease.
Collapse
Affiliation(s)
- Charalampos Papagoras
- 1st Department of Internal Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Paraskevi V Voulgari
- Rheumatology Clinic, Department of Internal Medicine, Medical School, University of Ioannina, Ioannina, Greece
| | - Alexandros A Drosos
- Rheumatology Clinic, Department of Internal Medicine, Medical School, University of Ioannina, Ioannina, Greece
| |
Collapse
|
10
|
Elbere I, Silamikelis I, Dindune II, Kalnina I, Ustinova M, Zaharenko L, Silamikele L, Rovite V, Gudra D, Konrade I, Sokolovska J, Pirags V, Klovins J. Baseline gut microbiome composition predicts metformin therapy short-term efficacy in newly diagnosed type 2 diabetes patients. PLoS One 2020; 15:e0241338. [PMID: 33125401 PMCID: PMC7598494 DOI: 10.1371/journal.pone.0241338] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023] Open
Abstract
Background The study was conducted to investigate the effects of metformin treatment on the human gut microbiome’s taxonomic and functional profile in the Latvian population, and to evaluate the correlation of these changes with therapeutic efficacy and tolerance. Methods In this longitudinal observational study, stool samples for shotgun metagenomic sequencing-based analysis were collected in two cohorts. The first cohort included 35 healthy nondiabetic individuals (metformin dose 2x850mg/day) at three time-points during metformin administration. The second cohort was composed of 50 newly-diagnosed type 2 diabetes patients (metformin dose–determined by an endocrinologist) at two concordant times. Patients were defined as Responders if their HbA1c levels during three months of metformin therapy had decreased by ≥12.6 mmol/mol (1%), while in Non-responders HbA1c were decreased by <12.6 mmol/mol (1%). Results Metformin reduced the alpha diversity of microbiota in healthy controls (p = 0.02) but not in T2D patients. At the species level, reduction in the abundance of Clostridium bartlettii and Barnesiella intestinihominis, as well as an increase in the abundance of Parabacteroides distasonis and Oscillibacter unclassified overlapped between both study groups. A large number of group-specific changes in taxonomic and functional profiles was observed. We identified an increased abundance of Prevotella copri (FDR = 0.01) in the Non-Responders subgroup, and enrichment of Enterococcus faecium, Lactococcus lactis, Odoribacter, and Dialister at baseline in the Responders group. Various taxonomic units were associated with the observed incidence of side effects in both cohorts. Conclusions Metformin effects are different in T2D patients and healthy individuals. Therapy induced changes in the composition of gut microbiome revealed possible mediators of observed short-term therapeutic effects. The baseline composition of the gut microbiome may influence metformin therapy efficacy and tolerance in T2D patients and could be used as a powerful prediction tool.
Collapse
Affiliation(s)
- Ilze Elbere
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | | | | | - Ineta Kalnina
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Monta Ustinova
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | | | | | - Vita Rovite
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Dita Gudra
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Ilze Konrade
- Latvian Biomedical Research and Study Centre, Riga, Latvia
- Riga Stradins University, Riga, Latvia
| | | | - Valdis Pirags
- Latvian Biomedical Research and Study Centre, Riga, Latvia
- Faculty of Medicine, University of Latvia, Riga, Latvia
| | - Janis Klovins
- Latvian Biomedical Research and Study Centre, Riga, Latvia
- * E-mail:
| |
Collapse
|
11
|
Castro Rocha FA, Duarte-Monteiro AM, Henrique da Mota LM, Matias Dinelly Pinto AC, Fonseca JE. Microbes, helminths, and rheumatic diseases. Best Pract Res Clin Rheumatol 2020; 34:101528. [PMID: 32448639 PMCID: PMC7203059 DOI: 10.1016/j.berh.2020.101528] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
There has been a progressive interest on modifications of the human defense system following insults occurring in the interface between our body and the external environment, as they may provoke or worsen disease states. Studies suggest that billions of germs, which compose the gut microbiota influence one's innate and adaptive immune responses at the intestinal level, but these microorganisms may also impact rheumatic diseases. The microbiota of the skin, respiratory, and urinary tracts may also be relevant in rheumatology. Evidence indicates that changes in the gut microbiome alter the pathogenesis of immune-mediated diseases such as rheumatoid arthritis and ankylosing spondylitis but also of other disorders like atherosclerosis and osteoarthritis. Therapeutic strategies to modify the microbiota, including probiotics and fecal microbiota transplantation, have been received with skepticism, which, in turn, has drawn attention back to previously developed interventions such as antibiotics. Helminths adapted to humans over the evolution process, but their role in disease modulation, particularly immune-mediated diseases, remains to be understood. The present review focuses on data concerning modifications of the immune system induced by interactions with microbes and pluricellular organisms, namely helminths, and their impact on rheumatic diseases. Practical aspects, including specific microbiota-targeted therapies, are also discussed.
Collapse
Affiliation(s)
- Francisco Airton Castro Rocha
- Departamento de Medicina Clínica, Liga de Reumatologia e Doenças Autoimunes, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brazil.
| | - Ana Margarida Duarte-Monteiro
- Serviço de Reumatologia e Doenças Ósseas Metabólicas, Hospital de Santa Maria, CHULN and Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisboa, Portugal
| | - Licia Maria Henrique da Mota
- Hospital Universitário de Brasília, Programa de Pós-graduação em Ciências Médicas, Faculdade de Medicina, Universidade de Brasília, Brazil
| | - Ana Carolina Matias Dinelly Pinto
- Departamento de Medicina Clínica, Liga de Reumatologia e Doenças Autoimunes, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - João Eurico Fonseca
- Serviço de Reumatologia e Doenças Ósseas Metabólicas, Hospital de Santa Maria, CHULN and Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisboa, Portugal
| |
Collapse
|
12
|
Rossini M, Epis OM, Tinazzi I, Grembiale RD, Iagnocco A. Role of the IL-23 pathway in the pathogenesis and treatment of enthesitis in psoriatic arthritis. Expert Opin Biol Ther 2020; 20:787-798. [PMID: 32129102 DOI: 10.1080/14712598.2020.1737855] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Enthesitis is a key feature of spondyloarthritis (SpA). Several studies have underlined the role of interleukin (IL)-23 in SpA development as a crucial cytokine in the pathogenesis of enthesitis. AREA COVERED This review summarizes recent evidence of the role of IL-23 in the pathogenesis of and as a target of the treatment of enthesitis. We review the definition, diagnosis and clinical impact of enthesitis and its connection with microbial infections, gut dysbiosis, and mechanical stress. We also review clinical trials and real-life studies of drugs targeting the p19 or p40 subunits of IL-23. EXPERT OPINION Novel therapies targeting the p19 or p40 subunit of IL-23 appear to be promising treatment options for patients with enthesitis. Although we are currently unable to identify the best therapeutic window to target IL-23 in SpA disease evolution, the promising ability of this therapy to control the gut-entheseal axis is increasing our knowledge of SpA pathogenesis.
Collapse
Affiliation(s)
- Maurizio Rossini
- Rheumatology Section, Department of Medicine, University of Verona , Verona, Italy
| | | | - Ilaria Tinazzi
- Unit of Rheumatology, IRCCS Sacro Cuore Don Calabria Hospital , Verona, Italy
| | | | - Annamaria Iagnocco
- Academic Rheumatology Centre, Università Degli Studi Di Torino , Turin, Italy
| |
Collapse
|
13
|
Defining the phenotype, pathogenesis and treatment of Crohn's disease associated spondyloarthritis. J Gastroenterol 2020; 55:667-678. [PMID: 32367294 PMCID: PMC7297835 DOI: 10.1007/s00535-020-01692-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 04/14/2020] [Indexed: 02/04/2023]
Abstract
Peripheral and axial spondyloarthritis are the most common extra-intestinal manifestations reported in patients with Crohn's disease. Despite the frequency of Crohn's disease associated spondyloarthritis, clinical diagnostic tools are variably applied in these cohorts and further characterization with validated spondyloarthritis disease activity indexes are needed. In addition, the pathogenesis of Crohn's disease associated spondyloarthritis is not well understood. Evidence of shared genetic, cellular, and microbial mechanisms underlying both Crohn's disease and spondyloarthritis highlight the potential for a distinct clinicopathologic entity. Existing treatment paradigms for Crohn's disease associated spondyloarthritis focus on symptom control and management of luminal inflammation. A better understanding of the underlying pathogenic mechanisms in Crohn's disease associated spondyloarthritis and the link between the gut microbiome and systemic immunity will help pave the way for more targeted and effective therapies. This review highlights recent work that has provided a framework for clinical characterization and pathogenesis of Crohn's disease associated spondyloarthritis and helps identify critical gaps that will help shape treatment paradigms.
Collapse
|
14
|
Choi RY, Asquith M, Rosenbaum JT. Fecal transplants in spondyloarthritis and uveitis: ready for a clinical trial? Curr Opin Rheumatol 2019. [PMID: 29538010 DOI: 10.1097/bor.0000000000000506] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW The intestinal microbiome is thought to play a role in the pathogenesis of inflammatory bowel disease (IBD). There are many shared clinical manifestations between IBD and spondyloarthritis (SpA), of which the most common are peripheral arthritis and uveitis. Clinical overlap along with similar genetics between these diseases suggests a possible shared pathogenetic mechanism, which might center on the intestinal microbiota. In this review, we discuss the available evidence that SpA is a microbiome-driven disease and indicate how SpA-associated uveitis could be tied to gut dysbiosis. We conclude by discussing different treatment paradigms targeting the intestinal microbiome for SpA. RECENT FINDINGS Recent studies support the growing evidence of the intestinal microbiome as a crucial player in SpA disease pathogenesis. There is emerging evidence that the gut microbiome may play a causative role in uveitis. SUMMARY The field is beginning to discover a new level of understanding how the intestinal microbiome is involved in SpA. Treatment methods to alter intestinal microbiota to treat SpA-related diseases are still in its infancy.
Collapse
Affiliation(s)
- Rene Y Choi
- Department of Ophthalmology, Casey Eye Institute
| | - Mark Asquith
- Division of Arthritis and Rheumatic Diseases, Department of Ophthalmology, Oregon Health and Science University
| | - James T Rosenbaum
- Department of Ophthalmology, Casey Eye Institute.,Division of Arthritis and Rheumatic Diseases, Department of Ophthalmology, Oregon Health and Science University.,Legacy Devers Eye Institute, Portland, Oregon, USA
| |
Collapse
|
15
|
Coit P, Kaushik P, Caplan L, Kerr GS, Walsh JA, Dubreuil M, Reimold A, Sawalha AH. Genome-wide DNA methylation analysis in ankylosing spondylitis identifies HLA-B*27 dependent and independent DNA methylation changes in whole blood. J Autoimmun 2019; 102:126-132. [PMID: 31128893 DOI: 10.1016/j.jaut.2019.04.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/28/2019] [Accepted: 04/29/2019] [Indexed: 01/19/2023]
Abstract
BACKGROUND AND OBJECTIVE Ankylosing spondylitis is a chronic inflammatory disease characterized by inflammation of the sacroiliac joints and the spine that can lead to significant pain, immobility, and disability. The etiology and pathogenesis of ankylosing spondylitis are incompletely understood, though most patients carry the HLA-B*27 allele. The objective of this study was to evaluate DNA methylation changes in ankylosing spondylitis with the goal of revealing novel mechanistic insights into this disease. METHODS Genome-wide DNA methylation analysis was performed in whole blood DNA samples using the Infinium MethylationEPIC array in patients with ankylosing spondylitis compared to age, sex, and race matched patients with osteoarthritis as a non-inflammatory disease control. We studied 24 patients with ankylosing spondylitis, including 12 patients who carry HLA-B*27 and 12 patients who are HLA-B*27 negative. DNA methylation analysis was performed with adjustment for blood cell composition in each sample. RESULTS We identified a total of 67 differentially methylated sites between ankylosing spondylitis patients and osteoarthritis controls. Hypermethylated genes found included GTPase-related genes, while hypomethylated genes included HCP5, which encodes a lncRNA within the MHC region, previously associated with genetic risk for psoriasis and toxic epidermal necrolysis. Carrying HLA-B*27 was associated with robust hypomethylation of HCP5, tubulin folding cofactor A (TBCA) and phospholipase D Family Member 6 (PLD6) in ankylosing spondylitis patients. Hypomethylation within HCP5 involves a CpG site that contains a single nucleotide polymorphism in linkage disequilibrium with HLA-B*27 and that controls DNA methylation at this locus in an allele-specific manner. CONCLUSIONS A genome-wide DNA methylation analysis in ankylosing spondylitis identified DNA methylation patterns that could provide potential novel insights into this disease. Our findings suggest that HLA-B*27 might play a role in ankylosing spondylitis in part through inducing epigenetic dysregulation.
Collapse
Affiliation(s)
- Patrick Coit
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Prashant Kaushik
- Rheumatology Service, Stratton VA Medical Center, Albany, NY, USA
| | - Liron Caplan
- Division of Rheumatology, Denver VA Medical Center and University of Colorado, Aurora, CO, USA
| | - Gail S Kerr
- Division of Rheumatology, Washington DC VAMC and Georgetown and Howard University, Washington, DC, USA
| | - Jessica A Walsh
- Division of Rheumatology, George E. Wahlen VA Medical Center & University of Utah, Salt Lake City, UT, USA
| | - Maureen Dubreuil
- Rheumatology, VA Boston Healthcare System & Boston University School of Medicine, Boston, MA, USA
| | - Andreas Reimold
- Division of Rheumatology, Dallas VA Medical Center and University of Texas - Southwestern, Dallas, TX, USA
| | - Amr H Sawalha
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Division of Rheumatology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA; Division of Rheumatology, Department of Medicine & Lupus Center of Excellence, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
16
|
Cristea D, Trandafir M, Bojinca VC, Ciontea AS, Andrei MM, Popa A, Lixandru BE, Militaru CM, Nascutiu AM, Predeteanu D, Ionescu R, Popescu C, Cotar AI, Popa MI, Spandidos DA, Codita I. Usefulness of complex bacteriological and serological analysis in patients with spondyloarthritis. Exp Ther Med 2019; 17:3465-3476. [PMID: 30988725 PMCID: PMC6447817 DOI: 10.3892/etm.2019.7336] [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: 07/25/2018] [Accepted: 02/14/2019] [Indexed: 12/17/2022] Open
Abstract
Spondyloarthritis (SpA) is a group of associated chronic systemic inflammatory immune-mediated rheumatic diseases affecting axial and peripheral joints and entheses. The aim of the present study was to identify what parameters are useful to determine in order to better understand the correlation between the disease activity/severity and the microbiological results/immune status against intestinal and/or urogenital pathogens. Microorganisms known to trigger SpA, including Klebsiella spp., Yersinia spp., Salmonella spp., Campylobacter spp. and Chlamydia spp., were analyzed in various specimens (stool, urine, synovial fluid and serum) collected from 27 randomly selected SpA patients and 26 healthy controls using a combined direct and indirect approach relying on conventional culture technique and nucleic acid-based assays together with serological testing by ELISA. Although Escherichia coli derived from phylogroup A prevailed in the gut microflora of the patients and controls, differences were observed regarding the representatives of the other phylogroups with a higher prevalence of E.coli members of phylogenetic group B1 in the stool specimens of patients. Antibodies against the targeted species were detected in SpA patients and controls, and the serological profiles of the former were more diverse and complex. In conclusion, the detection of anti-bacterial antibodies combined with other specific laboratory investigations should be more extensively used to monitor SpA patients in association with their symptoms and in order to determine and administer more effective therapeutics.
Collapse
Affiliation(s)
- Daniela Cristea
- 'Cantacuzino' National Medico-Military Institute for Research and Development, Bucharest 0050096, Romania
| | - Marius Trandafir
- 'Carol Davila' University of Medicine and Pharmacy, Bucharest 050474, Romania.,Department of Internal Medicine and Rheumatology, Hospital Sfanta Maria, Bucharest 011172, Romania
| | - Violeta Claudia Bojinca
- 'Carol Davila' University of Medicine and Pharmacy, Bucharest 050474, Romania.,Department of Internal Medicine and Rheumatology, Hospital Sfanta Maria, Bucharest 011172, Romania
| | - Adriana Simona Ciontea
- 'Cantacuzino' National Medico-Military Institute for Research and Development, Bucharest 0050096, Romania
| | - Melania Mihaela Andrei
- 'Cantacuzino' National Medico-Military Institute for Research and Development, Bucharest 0050096, Romania
| | - Andrei Popa
- 'Cantacuzino' National Medico-Military Institute for Research and Development, Bucharest 0050096, Romania
| | - Brandusa Elena Lixandru
- 'Cantacuzino' National Medico-Military Institute for Research and Development, Bucharest 0050096, Romania
| | - Cornelia Madalina Militaru
- 'Cantacuzino' National Medico-Military Institute for Research and Development, Bucharest 0050096, Romania
| | - Alexandra Maria Nascutiu
- 'Cantacuzino' National Medico-Military Institute for Research and Development, Bucharest 0050096, Romania.,'Carol Davila' University of Medicine and Pharmacy, Bucharest 050474, Romania
| | - Denisa Predeteanu
- Department of Internal Medicine and Rheumatology, Hospital Sfanta Maria, Bucharest 011172, Romania
| | - Ruxandra Ionescu
- 'Carol Davila' University of Medicine and Pharmacy, Bucharest 050474, Romania.,Department of Internal Medicine and Rheumatology, Hospital Sfanta Maria, Bucharest 011172, Romania
| | - Claudiu Popescu
- 'Carol Davila' University of Medicine and Pharmacy, Bucharest 050474, Romania.,'Dr Ion Stoia' Clinical Center for Rheumatic Diseases, Bucharest 030167, Romania
| | - Ani Ioana Cotar
- 'Cantacuzino' National Medico-Military Institute for Research and Development, Bucharest 0050096, Romania
| | - Mircea Ioan Popa
- 'Cantacuzino' National Medico-Military Institute for Research and Development, Bucharest 0050096, Romania.,'Carol Davila' University of Medicine and Pharmacy, Bucharest 050474, Romania
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Faculty of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Irina Codita
- 'Cantacuzino' National Medico-Military Institute for Research and Development, Bucharest 0050096, Romania.,'Carol Davila' University of Medicine and Pharmacy, Bucharest 050474, Romania
| |
Collapse
|
17
|
Niu Q, Wei W, Huang Z, Zhang J, Yang B, Wang L. Association between food allergy and ankylosing spondylitis: An observational study. Medicine (Baltimore) 2019; 98:e14421. [PMID: 30732197 PMCID: PMC6380781 DOI: 10.1097/md.0000000000014421] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Food allergies can alter the gut microbiome composition, increasing the risk of conditions such as ankylosing spondylitis (AS).To identify the association between specific allergens and AS, we investigated the differences in the serum levels of 14 food antigen-specific immunoglobulin (Ig) G between AS patients and healthy participants. The association between the levels of these antibodies and disease activity was assessed by measuring the inflammatory marker C-reactive protein (CRP).We enrolled 75 AS patients and 78 healthy controls who had undergone antigen-specific IgG tests in West China Hospital between January 2015 and October 2017, and performed enzyme-linked immunosorbent assays for specific IgG against 14 food allergens: rice, egg, mushroom, milk, pork, chicken, beef, crab, codfish, corn, soybean, tomato, shrimp, and wheat. The following tests were used to analyze differences between AS patients and healthy controls: χ test for sex, and a 2-tailed Student t-test or Mann-Whitney U test based on the results of Levene test for age and IgG levels. Correlations between IgG and CRP levels were calculated using a Spearman's correlation.AS patients had significantly higher serum levels of beef-, crab-, and pork-specific IgG than did healthy participants. In addition, the serum levels of pork-specific IgG were significantly and positively correlated with CRP.These results suggest that α-Gal, the predominant natural antigen in mammalian red meat, might play a potential role in the pathogenesis of AS, and therefore, AS patients should exclude such allergenic foods, including beef, crab and pork, from their daily diet.
Collapse
Affiliation(s)
- Qian Niu
- Department of Laboratory Medicine, West China Hospital
| | - Wei Wei
- West China Medical School, Sichuan University, Chengdu, China
| | | | - Junlong Zhang
- Department of Laboratory Medicine, West China Hospital
| | - Bin Yang
- Department of Laboratory Medicine, West China Hospital
| | - Lanlan Wang
- Department of Laboratory Medicine, West China Hospital
| |
Collapse
|
18
|
Olofsson T, Lindqvist E, Mogard E, Andréasson K, Marsal J, Geijer M, Kristensen LE, Wallman JK. Elevated faecal calprotectin is linked to worse disease status in axial spondyloarthritis: results from the SPARTAKUS cohort. Rheumatology (Oxford) 2019; 58:1176-1187. [DOI: 10.1093/rheumatology/key427] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 11/17/2018] [Indexed: 01/08/2023] Open
Affiliation(s)
- Tor Olofsson
- Department of Clinical Sciences Lund, Rheumatology, Lund University, Lund
- Department of Rheumatology, Skåne University Hospital, Lund
| | - Elisabet Lindqvist
- Department of Clinical Sciences Lund, Rheumatology, Lund University, Lund
- Department of Rheumatology, Skåne University Hospital, Lund
| | - Elisabeth Mogard
- Department of Clinical Sciences Lund, Rheumatology, Lund University, Lund
- Department of Rheumatology, Skåne University Hospital, Lund
| | - Kristofer Andréasson
- Department of Clinical Sciences Lund, Rheumatology, Lund University, Lund
- Department of Rheumatology, Skåne University Hospital, Lund
| | - Jan Marsal
- Department of Clinical Sciences Lund, Gastroenterology, Lund University, Lund
- Department of Gastroenterology, Skåne University Hospital, Lund
| | - Mats Geijer
- Department of Clinical Sciences Lund, Diagnostic Radiology, Lund University, Lund
- Department of Radiology, University of Gothenburg, Sahlgrenska Academy, Gothenburg, Sweden
| | - Lars Erik Kristensen
- Department of Clinical Sciences Lund, Rheumatology, Lund University, Lund
- Department of Rheumatology, The Parker Institute, Copenhagen University Hospital, Frederiksberg and Bispebjerg, Copenhagen, Denmark
| | - Johan K Wallman
- Department of Clinical Sciences Lund, Rheumatology, Lund University, Lund
- Department of Rheumatology, Skåne University Hospital, Lund
| |
Collapse
|
19
|
Kwan YH, Fong W, Ang XL, Tan CS, Tai BC, Huang Y, Bilger M, Phang JK, Tan HC, Lee JV, Sun L, Tan CT, Dong BQ, Koh HL, Leung YY, Lui NL, Yeo SI, Ng SC, Fong KY, Thumboo J, Østbye T. Traditional Chinese medicine (TCM) collaborative care for patients with axial spondyloarthritis (AcuSpA): protocol for a pragmatic randomized controlled trial. Trials 2019; 20:46. [PMID: 30642381 PMCID: PMC6332567 DOI: 10.1186/s13063-018-3117-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/08/2018] [Indexed: 11/24/2022] Open
Abstract
Background Axial spondyloarthritis (AxSpA) is a chronic disease which results in fatigue, pain, and reduced quality of life (QoL). Traditional Chinese medicine (TCM), especially acupuncture, has shown promise in managing pain. Although a TCM collaborative model of care (TCMCMC) has been studied in cancer, there are no randomized controlled trials investigating TCM in AxSpA. Therefore, we will conduct a pragmatic trial to determine the clinical effectiveness, safety, and cost-effectiveness of TCMCMC for patients with AxSpA. We define TCMCMC as standard TCM history taking and physical examination, acupuncture, and TCM non-pharmacological advice and communications with rheumatologists in addition to usual rheumatologic care. The purpose of this paper is to describe the rationale for and methodology of this trial. Methods/design This pragmatic randomized controlled trial will recruit 160 patients who are diagnosed with AxSpA and have inadequate response to non-steroidal anti-inflammatory drugs (NSAIDs). Simple randomization to usual rheumatologic care or the intervention (TCMCMC) with a 1:1 allocation ratio will be used. Ten 30-min acupuncture sessions will be provided to patients assigned to the TCMCMC arm. All participants will continue to receive usual rheumatologic care. The primary endpoint — spinal pain — will be evaluated at week 6. Secondary endpoints include clinical, quality of life, and economic outcome measures. Patients will be followed up for up to 52 weeks, and adverse events will be documented. Discussion This trial may provide evidence regarding the clinical effectiveness, safety, and cost-effectiveness of a TCMCMC for patients with AxSpA. Trial registration ClinicalTrials.gov, NCT03420404. Registered on 14 February 2018. Electronic supplementary material The online version of this article (10.1186/s13063-018-3117-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yu Heng Kwan
- Program in Health Services and Systems Research, Duke-NUS Medical School, Singapore, Singapore
| | - Warren Fong
- Department of Rheumatology and Immunology, Singapore General Hospital, Academia Building, Level 4, 20 College Road, Singapore, 169856, Singapore.,Duke-NUS Medical School, Singapore, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xiang Ling Ang
- Singapore Thong Chai Medical Institution, Singapore, Singapore
| | - Chuen Seng Tan
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Bee Choo Tai
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Youyi Huang
- Internal Medicine Residency, SingHealth, Singapore, Singapore
| | - Marcel Bilger
- Program in Health Services and Systems Research, Duke-NUS Medical School, Singapore, Singapore
| | - Jie Kie Phang
- Department of Rheumatology and Immunology, Singapore General Hospital, Academia Building, Level 4, 20 College Road, Singapore, 169856, Singapore
| | - Hui Chin Tan
- Singapore Thong Chai Medical Institution, Singapore, Singapore
| | - Jia Ven Lee
- Singapore Thong Chai Medical Institution, Singapore, Singapore
| | - Limin Sun
- Singapore Thong Chai Medical Institution, Singapore, Singapore
| | - Choy Tip Tan
- Singapore Thong Chai Medical Institution, Singapore, Singapore
| | - Bao Qiang Dong
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, People's Republic of China
| | - Hwee Ling Koh
- Department of Pharmacy, National University of Singapore, Singapore, Singapore
| | - Ying Ying Leung
- Department of Rheumatology and Immunology, Singapore General Hospital, Academia Building, Level 4, 20 College Road, Singapore, 169856, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Nai Lee Lui
- Department of Rheumatology and Immunology, Singapore General Hospital, Academia Building, Level 4, 20 College Road, Singapore, 169856, Singapore
| | - Siaw Ing Yeo
- Department of Rheumatology and Immunology, Singapore General Hospital, Academia Building, Level 4, 20 College Road, Singapore, 169856, Singapore
| | - Swee Cheng Ng
- Department of Rheumatology and Immunology, Singapore General Hospital, Academia Building, Level 4, 20 College Road, Singapore, 169856, Singapore
| | - Kok Yong Fong
- Department of Rheumatology and Immunology, Singapore General Hospital, Academia Building, Level 4, 20 College Road, Singapore, 169856, Singapore
| | - Julian Thumboo
- Program in Health Services and Systems Research, Duke-NUS Medical School, Singapore, Singapore. .,Department of Rheumatology and Immunology, Singapore General Hospital, Academia Building, Level 4, 20 College Road, Singapore, 169856, Singapore.
| | - Truls Østbye
- Program in Health Services and Systems Research, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
20
|
Vecellio M, Cohen CJ, Roberts AR, Wordsworth PB, Kenna TJ. RUNX3 and T-Bet in Immunopathogenesis of Ankylosing Spondylitis-Novel Targets for Therapy? Front Immunol 2019; 9:3132. [PMID: 30687330 PMCID: PMC6335330 DOI: 10.3389/fimmu.2018.03132] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 12/18/2018] [Indexed: 12/30/2022] Open
Abstract
Susceptibility to ankylosing spondylitis (AS) is polygenic with more than 100 genes identified to date. These include HLA-B27 and the aminopeptidases (ERAP1, ERAP2, and LNPEPS), which are involved in antigen processing and presentation to T-cells, and several genes (IL23R, IL6R, STAT3, JAK2, IL1R1/2, IL12B, and IL7R) involved in IL23 driven pathways of inflammation. AS is also strongly associated with polymorphisms in two transcription factors, RUNX3 and T-bet (encoded by TBX21), which are important in T-cell development and function. The influence of these genes on the pathogenesis of AS and their potential for identifying drug targets is discussed here.
Collapse
Affiliation(s)
- Matteo Vecellio
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom.,Oxford Musculoskeletal Biomedical Research Unit, National Institute for Health Research, Oxford, United Kingdom.,Oxford Comprehensive Biomedical Research Centre, Botnar Research Centre, National Institute for Health Research, Nuffield Orthopaedic Centre, Oxford, United Kingdom
| | - Carla J Cohen
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom.,Oxford Musculoskeletal Biomedical Research Unit, National Institute for Health Research, Oxford, United Kingdom.,Oxford Comprehensive Biomedical Research Centre, Botnar Research Centre, National Institute for Health Research, Nuffield Orthopaedic Centre, Oxford, United Kingdom
| | - Amity R Roberts
- Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Paul B Wordsworth
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom.,Oxford Musculoskeletal Biomedical Research Unit, National Institute for Health Research, Oxford, United Kingdom.,Oxford Comprehensive Biomedical Research Centre, Botnar Research Centre, National Institute for Health Research, Nuffield Orthopaedic Centre, Oxford, United Kingdom
| | - Tony J Kenna
- Translational Research Institute, Princess Alexandra Hospital, Brisbane, QLD, Australia.,Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
21
|
Leger AJ, Caspi RR. Visions of Eye Commensals: The Known and the Unknown About How the Microbiome Affects Eye Disease. Bioessays 2018; 40:e1800046. [PMID: 30289987 PMCID: PMC6354774 DOI: 10.1002/bies.201800046] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 08/13/2018] [Indexed: 12/13/2022]
Abstract
Until recently, the ocular surface is thought by many to be sterile and devoid of living microbes. It is now becoming clear that this may not be the case. Recent and sophisticated PCR analyses have shown that microbial DNA-based "signatures" are present within various ethnic, geographic, and contact lens wearing communities. Furthermore, using a mouse model of ocular surface disease, we have shown that the microbe, Corynebacterium mastitidis (C. mast), can stably colonize the ocular mucosa and that a causal relationship exists between ocular C. mast colonization and beneficial local immunity. While this constitutes proof-of-concept that a bona fide ocular microbiome that tunes immunity can exist at the ocular surface, there remain numerous unanswered questions to be addressed before microbiome-modulating therapies may be successfully developed. Here, the authors will briefly outline what is currently known about the local ocular microbiome as well as microbiomes associated with other sites, and how those sites may play a role in ocular surface immunity. Understanding how commensal microbes affect the ocular surface immune homeostasis has the potential revolutionize how we think about treating ocular surface disease.
Collapse
Affiliation(s)
- Anthony J. Leger
- Laboratory of Immunology National Eye Institute, Bethesda, MD 20892, USA; Department of Ophthalmology, University of Pittsburgh School of Medicine Pittsburgh, PA 15213, USA,
| | - Rachel R. Caspi
- Laboratory of Immunology National Eye Institute, Bethesda, MD 20892, USA,
| |
Collapse
|
22
|
Muhammad JS, Ghauri MI. Clinical patterns of seronegative spondyloarthropathies in a tertiary centre in Pakistan. J Taibah Univ Med Sci 2018; 13:298-301. [PMID: 31435337 PMCID: PMC6694949 DOI: 10.1016/j.jtumed.2018.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 03/03/2018] [Accepted: 03/06/2018] [Indexed: 11/18/2022] Open
Affiliation(s)
- Jibran Sualeh Muhammad
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Corresponding address: Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.
| | - Muhammad Ishaq Ghauri
- Department of Medicine, Jinnah Medical College Hospital, Jinnah Medical and Dental College, Karachi, Pakistan
| |
Collapse
|
23
|
Kragsnaes MS, Kjeldsen J, Horn HC, Munk HL, Pedersen FM, Holt HM, Pedersen JK, Holm DK, Glerup H, Andersen V, Fredberg U, Kristiansen K, Christensen R, Ellingsen T. Efficacy and safety of faecal microbiota transplantation in patients with psoriatic arthritis: protocol for a 6-month, double-blind, randomised, placebo-controlled trial. BMJ Open 2018; 8:e019231. [PMID: 29703851 PMCID: PMC5922473 DOI: 10.1136/bmjopen-2017-019231] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION An unbalanced intestinal microbiota may mediate activation of the inflammatory pathways seen in psoriatic arthritis (PsA). A randomised, placebo-controlled trial of faecal microbiota transplantation (FMT) infused into the small intestine of patients with PsA with active peripheral disease who are non-responsive to methotrexate (MTX) treatment will be conducted. The objective is to explore clinical aspects associated with FMT performed in patients with PsA. METHODS AND ANALYSIS This trial is a randomised, two-centre stratified, double-blind (patient, care provider and outcome assessor), placebo-controlled, parallel-group study. Eighty patients will be included and randomised (1:1) to either placebo (saline) or FMT provided from an anonymous healthy donor. Throughout the study, both groups will continue the weekly self-administered subcutaneous MTX treatment, remaining on the preinclusion dosage (15-25 mg/week). The clinical measures of psoriasis and PsA disease activity used include the Short (2-page) Health Assessment Questionnaire, the Dermatology Quality of Life Index, the Spondyloarthritis Research Consortium of Canada Enthesitis Index, the Psoriasis Area Severity Index, a dactylitis digit count, a swollen/tender joint count (66/68), plasma C reactive protein as well as visual analogue scales for pain, fatigue and patient and physician global assessments. The primary end point is the proportion of patients who experience treatment failure during the 6-month trial period. The number of adverse events will be registered throughout the study. ETHICS AND DISSEMINATION This is a proof-of-concept clinical trial and will be performed in agreement with Good Clinical Practice standards. Approvals have been obtained from the local Ethics Committee (DK-S-20150080) and the Danish Data Protection Agency (15/41684). The study has commenced in May 2017. Dissemination will be through presentations at national and international conferences and through publications in international peer-reviewed journal(s). TRIAL REGISTRATION NUMBER NCT03058900; Pre-results.
Collapse
Affiliation(s)
- Maja Skov Kragsnaes
- Department of Rheumatology, Odense University Hospital, Odense, Denmark
- Odense Patient data Explorative Network (OPEN), Department of Clinical Institute, University of Southern Denmark, Odense, Denmark
| | - Jens Kjeldsen
- Department of Gastroenterology, Odense University Hospital, Odense, Denmark
| | | | | | | | - Hanne Marie Holt
- Department of Clinical Microbiology, Odense University Hospital, Odense, Denmark
| | | | | | - Henning Glerup
- Diagnostic Centre, Silkeborg Regional Hospital, Silkeborg, Denmark
| | - Vibeke Andersen
- IRS-Centre Sonderjylland, Hospital of Southern Jutland, Aabenraa, Denmark
- Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Ulrich Fredberg
- Diagnostic Centre, Silkeborg Regional Hospital, Silkeborg, Denmark
| | - Karsten Kristiansen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
- Institute of Metagenomics, BGI-Shenzhen, Shenzhen, China
| | - Robin Christensen
- Musculoskeletal Statistics Unit, Parker Institute, Frederiksberg and Bispebjerg Hospital, Copenhagen, Denmark
| | - Torkell Ellingsen
- Department of Rheumatology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW The intestinal commensal microbiota are important in shaping immune cell repertoire and are influenced by host genetics. Because of this intricate interaction, an intestinal dysbiosis has been associated with multiple immune-mediated polygenic diseases. This review summarizes the literature on how alterations in the intestinal microbiota contribute to immune-mediated ocular disease, and how to potentially target the gut microbiome for therapeutic benefit. RECENT FINDINGS Several groups have demonstrated the importance of the intestinal microbiome in uveitis pathogenesis. Two groups showed that altering the microbiota with oral antibiotics results in reduced uveitis severity, and another group demonstrated that a commensal bacterial antigen activates retina-specific autoreactive T cells, potentially indicating a commensal trigger for uveitis. We have found that commensal bacterial metabolites, short chain fatty acids, can suppress autoimmune uveitis. Age-related macular degeneration is associated with an intestinal dysbiosis, which can be influenced by genetic risk alleles and age-related eye disease study (AREDS) supplementation. Strategies that might be effective for targeting the intestinal microbiota might involve several approaches, including the use of antibiotics, drugs that supplement beneficial bacterial components or target inflammatory bacterial strains, dietary strategies or microbial transplantation. SUMMARY The intestinal microbiota are potentially crucial in propagating inflammatory diseases of the eye, and can be targeted for therapeutic benefit.
Collapse
|
25
|
García-Kutzbach A, Chacón-Súchite J, García-Ferrer H, Iraheta I. Reactive arthritis: update 2018. Clin Rheumatol 2018; 37:869-874. [DOI: 10.1007/s10067-018-4022-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 01/30/2018] [Accepted: 02/01/2018] [Indexed: 01/08/2023]
|
26
|
Short chain fatty acids ameliorate immune-mediated uveitis partially by altering migration of lymphocytes from the intestine. Sci Rep 2017; 7:11745. [PMID: 28924192 PMCID: PMC5603543 DOI: 10.1038/s41598-017-12163-3] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 09/04/2017] [Indexed: 02/07/2023] Open
Abstract
Short chain fatty acids (SCFA) are metabolites of intestinal bacteria resulting from fermentation of dietary fiber. SCFA are protective in various animal models of inflammatory disease. We investigated the effects of exogenous administration of SFCAs, particularly propionate, on uveitis using an inducible model of experimental autoimmune uveitis (EAU). Oral SCFA administration attenuated uveitis severity in a mouse strain-dependent manner through regulatory T cell induction among lymphocytes in the intestinal lamina propria (LPL) and cervical lymph nodes (CLN). SCFA also suppressed effector T cell induction in the CLN and mesenteric lymph nodes (MLN). Alterations in intestinal morphology and gene expression demonstrated in the EAU model prior to the onset of uveitis were blunted by oral SCFA administration. Using a Kaede transgenic mouse, we demonstrated enhanced leukocyte trafficking between the intestine and the eye in EAU. Propionate suppressed T effector cell migration between the intestine and the spleen in EAU Kaede mice. In conclusion, our findings support exogenous administration of SCFAs as a potential treatment strategy for uveitis through the stabilization of subclinical intestinal alterations that occur in inflammatory diseases including uveitis, as well as prevention of trafficking of leukocytes between the gastrointestinal tract and extra-intestinal tissues.
Collapse
|
27
|
Abstract
Over the past 5 years, advances in high-throughput techniques and studies involving large cohorts of patients have led to considerable advances in the identification of novel genetic associations and immune pathways involved in ankylosing spondylitis (AS). These discoveries include genes encoding cytokine receptors, transcription factors, signalling molecules and transport proteins. Although progress has been made in understanding the functions and potential pathogenic roles of some of these molecules, much work remains to be done to comprehend their complex interactions and therapeutic potential in AS. In this Review, we outline the current knowledge of AS pathogenesis, including genetic risk associations, HLA-B27-mediated pathology, perturbations in antigen-presentation pathways and the contribution of the type 3 immune response.
Collapse
|
28
|
Abstract
Reactive arthritis is classified as a spondyloarthropathy. Current concepts of disease suggest an infectious trigger, followed by inflammatory arthritis. Several mechanisms have been proposed to explain the interaction of host susceptibility and microorganism. Diagnosis relies on a compatible clinical syndrome and microbiologic confirmation of the pathogen. Antibiotic therapy seems useful in Chlamydia-triggered arthritis. The role of antibiotics in arthritis triggered by enteric pathogens is less clear. The role of tumor necrosis factor alpha inhibitors in therapy is evolving. Many patients have a course limited to a few months, but others experience extraarticular disease and more prolonged courses.
Collapse
Affiliation(s)
- Steven K Schmitt
- Section of Bone and Joint Infections, Department of Infectious Disease, Medicine Institute, Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, 9500 Euclid Avenue, Desk G-21, Cleveland, OH 44195, USA.
| |
Collapse
|
29
|
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
|
30
|
Carter JD, Hudson AP. Recent advances and future directions in understanding and treating Chlamydia-induced reactive arthritis. Expert Rev Clin Immunol 2016; 13:197-206. [PMID: 27627462 DOI: 10.1080/1744666x.2017.1233816] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Reactive arthritis (ReA) is an inflammatory disease that can follow gastrointestinal or genitourinary infections. The primary etiologic agent for post-venereal ReA is the bacterium Chlamydia trachomatis; its relative, C pneumoniae, has also been implicated in disease induction although to a lesser degree. Studies have indicated that the arthritis is elicited by chlamydiae infecting synovial tissue in an unusual biologic state designated persistence. We review clinical aspects, host-pathogen interactions, and treatments for the disease. Areas covered: We briefly discuss both the historic and,more extensively, the current medical literature describing ReA, and we provide a discussion of the biology of the chlamydiae as it relates to elicitation of the disease. A summary of clinical aspects of Chlamydia-induced ReA is included to give context for approaches to treatment of the arthritis. Expert commentary: Basic research into the biology and host-pathogen interactions characteristic of C trachomatis has provided a wealth of information that underlies our current understanding of the pathogenic processes occurring in the ReA synovium. Importantly, a promising approach to cure of the disease is at hand. However, both basic and clinical research into Chlamydia-induced ReA has lagged over the last 5 years, including required studies relating to cure of the disease.
Collapse
Affiliation(s)
- John D Carter
- a Department of Internal Medicine, Division of Rheumatology , University of South Florida School of Medicine , Tampa , FL , USA
| | - Alan P Hudson
- b Department of Immunology and Microbiology , Wayne State University School of Medicine , Detroit , MI , USA
| |
Collapse
|
31
|
Rosenbaum JT, Asquith MJ. The Microbiome: a Revolution in Treatment for Rheumatic Diseases? Curr Rheumatol Rep 2016; 18:62. [DOI: 10.1007/s11926-016-0614-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|