1
|
Pellico MR, Day J, Shah M, Yi BY, Saketkoo LA, Lood C, Gupta L. Biorepositories For Global Rare Disease Research: A Narrative Review. Curr Rheumatol Rep 2025; 27:24. [PMID: 40397074 PMCID: PMC12095354 DOI: 10.1007/s11926-025-01189-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2025] [Indexed: 05/22/2025]
Abstract
PURPOSE OF THIS REVIEW Rare diseases, although individually infrequent, collectively impact a substantial number of people. Collaborative translational research using biospecimens is essential for advancing our understanding of the diverse characteristics and pathophysiology of rare diseases. Biobanks play a pivotal role in this endeavor by collecting, processing, transporting, and storing biospecimens, thereby serving as invaluable resources for medical research. In this review, we explore currently available biobanks, with a specific focus on those dedicated to rare rheumatic diseases. We also examine accessible best practice guidelines for establishing and maintaining high-quality biobanks, discuss the limitations and propose future directions for enhancing biobanking efforts in rare disease research. RECENT FINDINGS Advances in molecular and genomic technologies have expanded the role of biobanks, enhancing biomarker discovery and precision medicine. However, despite growth in biobanking capabilities, key challenges persist concerning ethics, interoperability, and biospecimen exchange, prompting active responses by various regulatory and governing bodies. Biobanking has transformed rare disease research. Strengthening national and international collaborations is essential for driving progress in this field and accelerating the development of novel therapeutic and precision medicine approaches.
Collapse
Affiliation(s)
- Maria Rosa Pellico
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Division of Rheumatology, ASST Gaetani Pini-CTO, Milan, Italy
| | - Jessica Day
- Department of Rheumatology, Royal Melbourne Hospital, Parkville, Australia
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Meera Shah
- Department of Rheumatology, Indraprastha Apollo Hospital, New Delhi, India
| | - Belina Y Yi
- Division of Pediatric Allergy, Immunology, and Rheumatology, Johns Hopkins University, Baltimore, MD, USA
| | - Lesley Ann Saketkoo
- New Orleans Scleroderma and Sarcoidosis Patient Care and Research Center, New Orleans, LA, USA
- Myositis International Health & Research Collaborative Alliance (MIHRA), Baltimore, USA
- LSU and Tulane University Schools of Medicine, New Orleans, LA, USA
| | - Christian Lood
- Division of Rheumatology, University of Washington, Seattle, WA, USA
| | - Latika Gupta
- Department of Rheumatology, Royal Wolverhampton Hospitals NHS Trust, Wolverhampton, UK.
- School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, UK.
- Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Centre for Musculoskeletal Research, The University of Manchester, Manchester, UK.
- Francis Crick Institute, London, UK.
| |
Collapse
|
2
|
Kamalanathan AS, Agarwal V, Talamini L, Muller S. Autophagy in myositis, a dysregulated pathway, and a target for therapy. Autoimmun Rev 2025; 24:103817. [PMID: 40262692 DOI: 10.1016/j.autrev.2025.103817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/27/2025] [Accepted: 04/14/2025] [Indexed: 04/24/2025]
Abstract
Corticosteroids and immunosuppressants are the mainstay of therapy for idiopathic inflammatory myopathies (IIMs). However, a significant therapeutic challenge extends beyond mitigating inflammation with these agents in achieving meaningful improvements in muscle strength and physical function, a goal that remains largely unmet. IIMs encompass a heterogeneous group of autoimmune disorders, including dermatomyositis, polymyositis, necrotizing autoimmune myopathy, inclusion body myositis, and others, characterized by chronic muscle inflammation, progressive weakness, and fatigue. The etiology of IIMs remains poorly understood, though potential contributors include environmental triggers (e.g., infections, medications, or injury) and genetic predisposition. To advance the development of novel therapeutic strategies, it is critical to elucidate the dysfunctional molecular and cellular pathways underlying IIM pathogenesis. Among these, dysregulated autophagy pathways have emerged as a promising target for therapeutic intervention. Specifically, impairments in lysosomal autophagy and mitophagy have been implicated in IIMs, and modulating these processes through targeted regulatory mechanisms may offer therapeutic benefits. This review provides a comprehensive synthesis of clinical and biological features of IIMs, the current diagnostic approaches and emerging biomarkers, evaluates the utility of existing biomarkers, and examines the relevance of animal models in IIM research. Furthermore, we explore the role of autophagic dysregulation in disease pathogenesis and provide a critical appraisal of current treatment modalities. Finally, we highlight emerging therapeutic targets and regulatory molecules under investigation, with a particular focus on autophagy modulation. Notably, autophagy inhibitors represent a novel and potentially transformative therapeutic avenue for patients with IIMs, offering hope for improved clinical outcomes.
Collapse
Affiliation(s)
- A S Kamalanathan
- Centre for BioSeparation Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Vikas Agarwal
- Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, Uttar Pradesh, India
| | - Laura Talamini
- CNRS and Strasbourg University Unit Biotechnology and Cell signalling/Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France
| | - Sylviane Muller
- CNRS and Strasbourg University Unit Biotechnology and Cell signalling/Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France; University of Strasbourg Institute for Advanced Study (USIAS), Strasbourg, France.
| |
Collapse
|
3
|
Kim H. Juvenile Dermatomyositis: Updates in Pathogenesis and Biomarkers, Current Treatment, and Emerging Targeted Therapies. Paediatr Drugs 2025; 27:57-72. [PMID: 39425894 PMCID: PMC11774970 DOI: 10.1007/s40272-024-00658-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/26/2024] [Indexed: 10/21/2024]
Abstract
Juvenile dermatomyositis is a rare systemic inflammatory autoimmune disease involving muscle, skin, and vessels. Most patients do not fully respond to initial therapy, instead having a chronic refractory or polycyclic disease course. Pathogenesis is not completely understood, but immune cell dysregulation, particularly of B cells, mitochondrial dysfunction, changes in neutrophils and neutrophil extracellular traps (NETs), and increased type I and type II interferon (IFN) signaling have been described. There are limited randomized controlled trials of drugs in juvenile dermatomyositis (JDM), and treatment is largely based on lower-quality data such as case series, retrospective studies, and open-label prospective studies. These data have been compiled into expert recommendations or consensus treatment plans, which help guide therapy. While initial therapy is more standard with most including corticosteroids (high-dose oral and/or pulse intravenous methylprednisolone) and methotrexate, for refractory patients, guidelines are more varied with multiple options or combinations, including biologic therapies. There is a clear need for more efficacious and personalized therapy in JDM. Emerging treatment options worthy of further study in JDM include targeting IFN-signaling (JAK, IFNAR1, IFN beta), B-cells (CD20, CD19, BAFF, TACI, CD38, BCMA) including Chimeric Antigen Receptor (CAR)-T cell therapy, mitochondrial dysfunction, and NETs.
Collapse
Affiliation(s)
- Hanna Kim
- National Institute of Arthritis Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
4
|
Sparling AC, Ward JM, Sarkar K, Schiffenbauer A, Farhadi PN, Smith MA, Rahman S, Zerrouki K, Miller FW, Li JL, Casey KA, Rider LG. Neutrophil and mononuclear leukocyte pathways and upstream regulators revealed by serum proteomics of adult and juvenile dermatomyositis. Arthritis Res Ther 2024; 26:196. [PMID: 39529136 PMCID: PMC11552237 DOI: 10.1186/s13075-024-03421-7] [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: 05/09/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVES Serum protein abundance was assessed in adult and juvenile dermatomyositis (DM and JDM) patients to determine differentially regulated proteins, altered pathways, and candidate disease activity biomarkers. METHODS Serum protein expression from 17 active adult DM and JDM patients each was compared to matched, healthy control subjects by a multiplex immunoassay. Pathway analysis and protein clustering of the differentially regulated proteins were examined to assess underlying mechanisms. Candidate disease activity biomarkers were identified by correlating protein expression with disease activity measures. RESULTS Seventy-eight of 172 proteins were differentially expressed in the sera of DM and JDM patients compared to healthy controls. Forty-eight proteins were differentially expressed in DM, 32 proteins in JDM, and 14 proteins in both DM and JDM. Twelve additional differentially expressed proteins were identified after combining the DM and JDM cohorts. C-X-C motif chemokine ligand 10 (CXCL10) was the most strongly upregulated protein in both DM and JDM sera. Other highly upregulated proteins in DM included S100 calcium binding protein A12 (S100A12), CXCL9, and nicotinamide phosphoribosyltransferase (NAMPT), while highly upregulated proteins in JDM included matrix metallopeptidase 3 (MMP3), growth differentiation factor 15 (GDF15), and von Willebrand factor (vWF). Pathway analysis indicated that phosphoinositide 3-kinase (PI3K), p38 mitogen-activated protein kinase (MAPK), and toll-like receptor 7 (TLR7) signaling were activated in DM and JDM. Additional pathways specific to DM or JDM were identified. A protein cluster associated with neutrophils and mononuclear leukocytes and a cluster of interferon-associated proteins were observed in both DM and JDM. Twenty-two proteins in DM and 24 proteins in JDM sera correlated with global, muscle, and/or skin disease activity. Seven proteins correlated with disease activity measures in both DM and JDM sera. IL-1 receptor like 1 (IL1RL1) emerged as a candidate global disease activity biomarker in DM and JDM. CONCLUSION Coordinate analysis of protein expression in DM and JDM patient sera by a multiplex immunoassay validated previous gene expression studies and identified novel dysregulated proteins, altered signaling pathways, and candidate disease activity biomarkers. These findings may further inform the assessment of DM and JDM patients and aid in the identification of potential therapeutic targets.
Collapse
Affiliation(s)
- A Clare Sparling
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Building 10, CRC Rm 6-5700, MSC 1301 10 Center Drive, Bethesda, MD, 20892-1301, USA
| | - James M Ward
- Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, USA
| | - Kakali Sarkar
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Building 10, CRC Rm 6-5700, MSC 1301 10 Center Drive, Bethesda, MD, 20892-1301, USA
| | - Adam Schiffenbauer
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Building 10, CRC Rm 6-5700, MSC 1301 10 Center Drive, Bethesda, MD, 20892-1301, USA
| | - Payam Noroozi Farhadi
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Building 10, CRC Rm 6-5700, MSC 1301 10 Center Drive, Bethesda, MD, 20892-1301, USA
| | | | - Saifur Rahman
- BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | | | - Frederick W Miller
- Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, USA
| | - Jian-Liang Li
- Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, USA
| | - Kerry A Casey
- BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Lisa G Rider
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Building 10, CRC Rm 6-5700, MSC 1301 10 Center Drive, Bethesda, MD, 20892-1301, USA.
| |
Collapse
|
5
|
Tarvin SE, Sherman MA, Kim H, Balmuri N, Brown AG, Chow A, Gewanter HL, de Guzman MM, Huber AM, Kim S, Klein-Gitelman MS, Perron MM, Robinson AB, Sabbagh SE, Savani S, Shenoi S, Spitznagle J, Stingl C, Syverson G, Tory H, Spencer C. Childhood Arthritis and Rheumatology Research Alliance Biologic Disease-Modifying Antirheumatic Drug Consensus Treatment Plans for Refractory Moderately Severe Juvenile Dermatomyositis. Arthritis Care Res (Hoboken) 2024; 76:1532-1539. [PMID: 38937134 DOI: 10.1002/acr.25393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/04/2024] [Accepted: 06/13/2024] [Indexed: 06/29/2024]
Abstract
OBJECTIVE The objective was to develop consensus treatment plans (CTPs) for patients with refractory moderately severe juvenile dermatomyositis (JDM) treated with biologic disease-modifying antirheumatic drugs (bDMARDs). METHODS The Biologics Workgroup of the Childhood Arthritis and Rheumatology Research Alliance JDM Research Committee used case-based surveys, consensus framework, and nominal group technique to produce bDMARD CTPs for patients with refractory moderately severe JDM. RESULTS Four bDMARD CTPs were proposed: tumor necrosis factor α (TNFα) inhibitor (adalimumab or infliximab), abatacept, rituximab, and tocilizumab. Each CTP has different options for dosing and/or route. Among 76 respondents, consensus was achieved for the proposed CTPs (93% [67 of 72]) as well as for patient characteristics, assessments, outcome measures, and follow-up. By weighted average, respondents indicated that they would most likely administer rituximab, followed by abatacept, TNFα inhibitor, and tocilizumab. CONCLUSION CTPs for the administration of bDMARDs in refractory moderately severe JDM were developed using consensus methodology. The implementation of the bDMARD CTPs will lay the groundwork for registry-based prospective comparative effectiveness studies.
Collapse
Affiliation(s)
| | - Matthew A Sherman
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | - Hanna Kim
- National Institute of Arthritis Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland
| | | | - Amanda G Brown
- University of Arkansas for Medical Sciences/Arkansas Children's Hospital, Little Rock
| | - Albert Chow
- Loma Linda Children's Hospital, Loma Linda, California
| | - Harry L Gewanter
- Children's Hospital of Richmond at Virginia Commonwealth University Health
| | | | - Adam M Huber
- IWK Health, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Susan Kim
- University of California, San Francisco
| | - Marisa S Klein-Gitelman
- Ann & Robert Lurie Children's Hospital of Chicago and Northwestern University, Chicago, Illinois
| | | | | | | | - Sonia Savani
- Medical University of South Carolina, Charleston
| | - Susan Shenoi
- University of Washington and Seattle Children's Hospital and Research Center, Seattle
| | - Jacob Spitznagle
- University of Washington and Seattle Children's Hospital and Research Center, Seattle
| | | | | | - Heather Tory
- Connecticut Children's Medical Center, Hartford, and University of Connecticut School of Medicine, Farmington
| | - Charles Spencer
- Batson Children's Hospital, University of Mississippi Medical Center, Jackson
| |
Collapse
|
6
|
Condessa L, Dias S, Moura Antunes S, Martins M, Madureira I. When a Child Refuses to Play: A Rare Myopathy. Cureus 2024; 16:e68372. [PMID: 39355458 PMCID: PMC11443743 DOI: 10.7759/cureus.68372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2024] [Indexed: 10/03/2024] Open
Abstract
Idiopathic inflammatory myopathies (IIM) are a rare group of systemic diseases characterized by progressive proximal muscle weakness and skeletal muscle inflammation. We describe a clinical report of a seven-year-old boy presenting with myalgia and proximal muscle weakness beginning three weeks earlier, with laboratory, MRI, and muscle biopsy findings consistent with IIM. The patient was treated with corticosteroids, methotrexate, immunoglobulin, and intensive motor rehabilitation, with favorable evolution. Diagnosis of Juvenile Polymyositis was confirmed. Three years later, we assisted a relapse of muscle weakness and muscle cytolysis with the onset of bilateral eyelid skin microulcers compatible with dermatomyositis. This report intends to highlight the importance of early diagnosis and treatment in IIM due to the significant burden associated with this group of diseases. In this case, the late onset of the skin lesion contributed to the challenge in this diagnosis.
Collapse
Affiliation(s)
- Luzia Condessa
- Pediatrics, Hospital de Cascais Dr. José de Almeida, Lisbon, PRT
| | - Susana Dias
- Pediatrics, Hospital de Cascais Dr. José de Almeida, Lisbon, PRT
| | | | - Mafalda Martins
- Pediatrics, Hospital de Cascais Dr. José de Almeida, Lisbon, PRT
| | - Inês Madureira
- Pediatric Rheumatology, Hospital Dona Estefânia, Centro Hospitalar Universitário Lisboa Central, Lisboa, PRT
| |
Collapse
|
7
|
Valões CCM, Arabi TM, Braga ALF, Campos LMA, Aikawa NE, Kozu KT, Silva CA, Farhat SCL, Elias AM. The influence of environmental factors related to Juvenile Dermatomyositis (JDM), its course and refractoriness to treatment. Adv Rheumatol 2024; 64:64. [PMID: 39215374 DOI: 10.1186/s42358-024-00408-5] [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: 03/14/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVE To evaluate the influence of environmental factors and prematurity relating to juvenile dermatomyositis (JDM), its course and refractoriness to treatment. METHODS A case-control study with 35 patients followed up at a tertiary hospital and 124 healthy controls, all residents of São Paulo. Patients were classified according to monocyclic, polycyclic or chronic disease courses and refractoriness to treatment. The daily concentrations of pollutants (inhalable particulate matter-PM10, sulfur dioxide-SO2, nitrogen dioxide-NO2, ozone-O3 and carbon monoxide-CO) were provided by the Environmental Company of São Paulo. Data from the population were obtained through a questionnaire. RESULTS Fifteen patients had monocyclic courses, and 19 polycyclic/chronic courses. Eighteen patients were refractory to treatment. Maternal occupational exposure to inhalable agents (OR = 17.88; IC 95% 2.15-148.16, p = 0.01) and exposure to O3 in the fifth year of life (third tertile > 86.28µg/m3; OR = 6.53, IC95% 1.60-26.77, p = 0.01) were risk factors for JDM in the multivariate logistic regression model. The presence of a factory/quarry at a distance farther than 200 meters from daycare/school (OR = 0.22; IC 95% 0.06-0.77; p = 0.02) was a protective factor in the same analysis. Prematurity, exposure to air pollutants/cigarette smoke/sources of inhalable pollutants in the mother's places of residence and work during the gestational period were not associated with JDM. Prematurity, maternal exposure to occupational pollutants during pregnancy as well as patient's exposure to ground-level pollutants up to the fifth year of life were not associated with disease course and treatment refractoriness. CONCLUSION Risk factors for JDM were maternal occupational exposure and exposure to O3 in the fifth year of life.
Collapse
Affiliation(s)
- Clarissa C M Valões
- Hospital da Criança, Avenida Juca Sampaio, 67, Jacintinho Maceió, AL, 57042-530, Brazil.
| | - Tamima M Arabi
- Rheumatology Unit, Child and Adolescent Institute, Hospital das Clínicas, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Alfésio L F Braga
- Environmental Epidemiology Study Group, Laboratory of Experimental Air Pollution, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Lúcia M A Campos
- Rheumatology Unit, Child and Adolescent Institute, Hospital das Clínicas, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Nádia E Aikawa
- Rheumatology Unit, Child and Adolescent Institute and Rheumatology Division, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Kátia T Kozu
- Rheumatology Unit, Child and Adolescent Institute, Hospital das Clínicas, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Clovis A Silva
- Rheumatology Unit, Child and Adolescent Institute, Hospital das Clínicas, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Sylvia C L Farhat
- Environmental Epidemiology Study Group, Laboratory of Experimental Air Pollution, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Adriana M Elias
- Rheumatology Unit, Child and Adolescent Institute, Hospital das Clínicas, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
8
|
Kobayashi I. Advances in Juvenile Dermatomyositis: Pathophysiology, Diagnosis, Treatment and Interstitial Lung Diseases-A Narrative Review. CHILDREN (BASEL, SWITZERLAND) 2024; 11:1046. [PMID: 39334579 PMCID: PMC11430821 DOI: 10.3390/children11091046] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024]
Abstract
Juvenile idiopathic inflammatory myopathy (JIIM) is a rare systemic autoimmune disease characterized by skeletal muscle weakness with or without a skin rash. Juvenile dermatomyositis (JDM) is the most common subtype of JIIM, accounting for 80% of JIIM. Recent studies identified several myositis-specific autoantibodies (MSAs) and myositis-associated autoantibodies (MAAs). Each MSA or MAA is associated with distinct clinical features and outcomes, although there are several differences in the prevalence of MSA/MAA and autoantibody-phenotype relationships between age and ethnic groups. Histopathological studies have revealed critical roles of type I interferons and vasculopathy in the development of JDM. Serological classification mostly corresponds to clinicopathological classification. Novel therapeutic agents, such as biologics and Janus kinase inhibitors (JAKi), have been developed; however, to date, there is a lack of high-level evidence. As advances in treatment have reduced the mortality rate of JIIM, recent studies have focused on medium- and long-term outcomes. However, rapidly progressive interstitial lung disease (RP-ILD) remains a major cause of death in anti-melanoma differentiation gene 5 autoantibody-positive JDM. Early diagnosis and intervention using a multi-drug regimen is critical for the treatment of RP-ILD. Rituximab and JAKi may reduce mortality in patients with JDM-associated RP-ILD refractory to conventional therapy.
Collapse
Affiliation(s)
- Ichiro Kobayashi
- Center for Pediatric Allergy and Rheumatology, KKR Sapporo Medical Center, 3-40 Hiragishi 1-6, Toyohira-ku, Sapporo 060-0931, Japan
| |
Collapse
|
9
|
Ryan MA, Ermarth A. Inflammatory Causes of Dysphagia in Children. Otolaryngol Clin North Am 2024; 57:669-684. [PMID: 38637195 DOI: 10.1016/j.otc.2024.03.002] [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] [Indexed: 04/20/2024]
Abstract
Gastroesophageal reflux (GER) and eosinophilic esophagitis (EoE) are the most common inflammatory causes of pediatric dysphagia, but several other less prevalent conditions should be considered. These conditions can affect one or several aspects of the swallowing process. In some inflammatory conditions dysphagia may be an early symptom. Esophagoscopy and instrumental swallow studies are often needed to determine the underlying diagnosis and best treatment plan. In some inflammatory conditions dysphagia can portend a worse outcome and need for more aggressive treatment of the underlying condition. Consultations with speech language pathology, gastroenterology, dietetics, allergy/immunology and/or rheumatology are often needed to optimize management.
Collapse
Affiliation(s)
- Marisa A Ryan
- Pediatric Otolaryngology, Peak ENT Associates, 1055 North 300 West, Suite 401, Provo, UT 84604, USA.
| | - Anna Ermarth
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and Nutrition, University of Utah School of Medicine, 81 Mario Capecchi Drive, Salt Lake City, UT 84113, USA
| |
Collapse
|
10
|
Koester ST, Chow A, Pepper-Tunick E, Lee P, Eckert M, Brenchley L, Gardner P, Song HJ, Li N, Schiffenbauer A, Volochayev R, Bayat N, McLean JS, Rider LG, Shenoi S, Stevens AM, Dey N. Familial clustering of dysbiotic oral and fecal microbiomes in juvenile dermatomyositis. Sci Rep 2024; 14:16158. [PMID: 38997299 PMCID: PMC11245510 DOI: 10.1038/s41598-024-60225-0] [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: 03/15/2023] [Accepted: 04/19/2024] [Indexed: 07/14/2024] Open
Abstract
Juvenile dermatomyositis (JDM) is a rare immune-mediated disease of childhood with putative links to microbial exposures. In this multi-center, prospective, observational cohort study, we evaluated whether JDM is associated with discrete oral and gut microbiome signatures. We generated 16S rRNA sequencing data from fecal, saliva, supragingival, and subgingival plaque samples from JDM probands (n = 28). To control for genetic and environmental determinants of microbiome community structure, we also profiled microbiomes of unaffected family members (n = 27 siblings, n = 26 mothers, and n = 17 fathers). Sample type (oral-vs-fecal) and nuclear family unit were the predominant variables explaining variance in microbiome diversity, more so than having a diagnosis of JDM. The oral and gut microbiomes of JDM probands were more similar to their own unaffected siblings than they were to the microbiomes of other JDM probands. In a sibling-paired within-family analysis, several potentially immunomodulatory bacterial taxa were differentially abundant in the microbiomes of JDM probands compared to their unaffected siblings, including Faecalibacterium (gut) and Streptococcus (oral cavity). While microbiome features of JDM are often shared by unaffected family members, the loss or gain of specific fecal and oral bacteria may play a role in disease pathogenesis or be secondary to immune dysfunction in susceptible individuals.
Collapse
Affiliation(s)
- Sean T Koester
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- University of Kansas School of Medicine, Kansas City, USA
| | - Albert Chow
- Department of Pediatrics, Division of Rheumatology, University of Washington, Seattle, WA, USA
- Loma Linda University, Loma Linda, USA
| | - Evan Pepper-Tunick
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, USA
| | - Peggy Lee
- School of Dentistry, University of Washington, Seattle, WA, USA
| | - Mary Eckert
- Center for Clinical and Translational Research, Seattle Children's Research Institute, Seattle, WA, USA
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA
| | - Laurie Brenchley
- Office of the Clinical Director, NIDCR, National Institutes of Health, Bethesda, MD, USA
| | - Pamela Gardner
- Office of the Clinical Director, NIDCR, National Institutes of Health, Bethesda, MD, USA
- Oral Oncology at BC Cancer, Vancouver, BC, Canada
| | - Hyun Jung Song
- School of Dentistry, University of Washington, Seattle, WA, USA
| | - Naisi Li
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Adam Schiffenbauer
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Rita Volochayev
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Nastaran Bayat
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, MD, USA
- Social and Scientific Systems, Inc., A DLH Holdings Corp. Company, Silver Spring, MD, USA
| | - Jeffrey S McLean
- Department of Periodontics, University of Washington, Seattle, WA, USA
| | - Lisa G Rider
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Susan Shenoi
- Department of Pediatrics, Division of Rheumatology, University of Washington, Seattle, WA, USA
- Center for Clinical and Translational Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Anne M Stevens
- Department of Pediatrics, Division of Rheumatology, University of Washington, Seattle, WA, USA
- Center for Clinical and Translational Research, Seattle Children's Research Institute, Seattle, WA, USA
- Janssen, a Wholly Owned Subsidiary of Johnson & Johnson, Raritan, USA
| | - Neelendu Dey
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Medicine, Division of Gastroenterology, University of Washington, Seattle, WA, USA.
- Microbiome Research Initiative, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| |
Collapse
|
11
|
Sherman MA, Farhadi PN, Pak K, Trieu EP, Sarkar K, Targoff IN, Neely ML, Mammen AL, Rider LG, Childhood Myositis Heterogeneity Collaborative Study Group. Myositis-Associated Autoantibodies in Patients With Juvenile Myositis Are Associated With Refractory Disease and Mortality. Arthritis Rheumatol 2024; 76:963-972. [PMID: 38272842 PMCID: PMC11136598 DOI: 10.1002/art.42813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/08/2024] [Accepted: 01/22/2024] [Indexed: 01/27/2024]
Abstract
OBJECTIVE Myositis-associated autoantibodies (MAAs) have been associated with overlap myositis, certain disease manifestations such as interstitial lung disease (ILD), and worse prognosis in the idiopathic inflammatory myopathies. MAAs overall remain largely uncharacterized in patients with juvenile-onset myositis. Moreover, it is unknown whether the number of MAAs is associated with disease severity. METHODS Patients with juvenile myositis in cross-sectional natural history studies who underwent testing for myositis autoantibodies were included. Demographics, myositis autoantibodies, clinical characteristics, medications received, and outcomes of those with and without MAAs were compared. Multivariable logistic regression was performed to determine whether the number of MAAs detected was associated with severe disease features. RESULTS Among 551 patients, 36% had an MAA and 13% had more than one MAA. Among those who were MAA positive, there was a higher frequency of overlap myositis (18% vs 5.9%, P < 0.001). MAA positivity was associated with certain clinical features, including Raynaud phenomenon (odds ratio [OR] 2.44, 95% confidence interval [CI] 1.41-4.28) and ILD (OR 3.43, 95% CI 1.75-6.96), as well as a chronic disease course (OR 1.72, 95% CI 1.10-2.72) and mortality (OR 3.76, 95% CI 1.72-8.43). The number of MAAs was also associated with mortality (OR 1.83, 95% CI 1.16-2.86). CONCLUSION MAAs were prevalent in a large cohort of patients with juvenile myositis. ILD, refractory disease, and mortality were associated with MAA positivity. Prospective studies are needed to determine whether early detection of MAAs may lead to improved outcomes for patients with juvenile myositis.
Collapse
Affiliation(s)
- Matthew A. Sherman
- Muscle Disease Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Payam Noroozi Farhadi
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Katherine Pak
- Muscle Disease Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Edward P. Trieu
- Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Kakali Sarkar
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Ira N. Targoff
- Veterans Affairs Medical Center, University of Oklahoma Health Sciences Center, and Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Megan L. Neely
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Andrew L. Mammen
- Muscle Disease Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lisa G. Rider
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | | |
Collapse
|
12
|
Shurin MR, Wheeler SE. Clinical Significance of Uncommon, Non-Clinical, and Novel Autoantibodies. Immunotargets Ther 2024; 13:215-234. [PMID: 38686351 PMCID: PMC11057673 DOI: 10.2147/itt.s450184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 04/17/2024] [Indexed: 05/02/2024] Open
Abstract
Autoantibodies are a common mark of autoimmune reaction and their identification in the patients' serum, cerebrospinal fluid, or tissues is generally believed to represent diagnostic or prognostic biomarkers of autoimmune diseases or autoinflammatory conditions. Traditionally, autoantibody testing is an important part of the clinical examination of suspected patients, and in the absence of reliable T cell tests, characterization of autoantibody responses might be suitable in finding causes of specific autoimmune responses, their strength, and sometimes commencement of autoimmune disease. Autoantibodies are also useful for prognostic stratification in clinically diverse groups of patients if checked repeatedly. Antibody discoveries are continuing, with important consequences for verifying autoimmune mechanisms, diagnostic feasibility, and clinical management. Adding newly identified autoantibody-autoantigen pairs to common clinical laboratory panels should help upgrade and harmonize the identification of systemic autoimmune rheumatic disorders and other autoimmune conditions. Herein, we aim to summarize our current knowledge of uncommon and novel autoantibodies in the context of discussing their validation, diagnostic practicability, and clinical relevance. The regular updates within the field are important and well justified.
Collapse
Affiliation(s)
- Michael R Shurin
- Division of Clinical Immunopathology, Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Sarah E Wheeler
- Division of Clinical Immunopathology, Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
13
|
Sherman MA, Yang Q, Gutierrez-Alamillo L, Pak K, Flegel WA, Mammen AL, Rider LG, Casciola-Rosen LA, the Childhood Myositis Heterogeneity Collaborative Study Group. Clinical Features and Immunogenetic Risk Factors Associated With Additional Autoantibodies in Anti-Transcriptional Intermediary Factor 1γ Juvenile-Onset Dermatomyositis. Arthritis Rheumatol 2024; 76:631-637. [PMID: 38059274 PMCID: PMC10965375 DOI: 10.1002/art.42768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 11/20/2023] [Accepted: 11/28/2023] [Indexed: 12/08/2023]
Abstract
OBJECTIVE Novel autoantibody specificities including anti-CCAR1 were recently discovered in adult patients with anti-transcriptional intermediary factor (TIF1)-positive dermatomyositis (DM) and were associated with attenuated cancer emergence. The aims of the present study were to examine whether these autoantibodies occur in patients with juvenile-onset DM (JDM) and to determine their associated features. METHODS Sera from 150 patients with anti-TIF1γ autoantibody-positive JDM in a cross-sectional cohort and 90 juvenile healthy controls were assayed for anti-CCAR1, anti-C1Z1, anti-IMMT, anti-TBL1XR1, and anti-Sp4 autoantibodies. Demographics, myositis autoantibodies, clinical features, medications, outcomes, and HLA-DRB1 and HLA-DQA1 alleles were compared between those with and without these autoantibodies. RESULTS Any one of the anti-TIF1γ-associated autoantibodies was present in 44 patients (29%) overall, including 25 (17%) with anti-Sp4, 22 (15%) with anti-TBL1XR1, 14 (9%) with anti-CCAR1, 2 (1%) with anti-C1Z1, and 2 (1%) with anti-IMMT autoantibodies. These anti-TIF1γ-associated autoantibodies frequently co-occurred. Patients with any of the anti-TIF1γ-associated autoantibodies had less frequent falling (34% [15] vs. 53% [56], P = 0.032) and lower peak muscle enzymes. None of the patients had cancer. Among White patients, HLA-DRB1*03 was protective against an anti-TIF1γ-associated autoantibody (odds ratio 0.20, 95% confidence interval 0.07-0.52). CONCLUSION Autoantibodies associated with anti-TIF1γ were found in isolation and in combination among a subset of patients with JDM. Patients with these autoantibodies had less severe muscle disease and were not enriched for HLA-DRB1*03. Additional autoantibodies among patients with positive anti-TIF1γ with JDM likely contribute to the heterogeneity of the anti-TIF1γ serologic subgroup.
Collapse
Affiliation(s)
- Matthew A. Sherman
- Muscle Disease Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Qingyuan Yang
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Katherine Pak
- Muscle Disease Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Willy A. Flegel
- Department of Transfusion Medicine, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Andrew L. Mammen
- Muscle Disease Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lisa G. Rider
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Livia A. Casciola-Rosen
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | | |
Collapse
|
14
|
Raupov R, Suspitsin E, Preobrazhenskaya EV, Kostik M. Interferon type I signature associated with skin disease in juvenile dermatomyositis. Front Med (Lausanne) 2024; 11:1214920. [PMID: 38420360 PMCID: PMC10899462 DOI: 10.3389/fmed.2024.1214920] [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: 04/30/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
Background Interferon type I (IFN-I) signaling system hyperactivation plays an important role in the pathogenesis of juvenile dermatomyositis (JDM). Aim of the study To analyze IFN-I score with disease activity in patients with JDM. Materials and methods Clinical manifestations laboratory data, and treatment options were analyzed in 15 children with JDM. Disease activity was assessed by CMAS (childhood myositis assessment tool) and CAT (cutaneous assessment tool) scores. IFN I-score was assessed by RT-PCR quantitation of 5 IFN I-regulated transcripts (IFI44L, IFI44, IFIT3, LY6E, MXA1). Results All patients had skin and muscle involvement, some had a fever (n = 8), swallowing disorders (n = 4), arthritis (n = 5), calcinosis (n = 3), lipodystrophy (n = 2), and interstitial lung disease (n = 5). Twelve patients had elevated IFN I-score and it was correlated with skin disease activity. Ten patients had clinically active disease and the level of IFN I-score and its components were higher than in patients with inactive disease (8.8 vs. 4.2, p = 0.011). IFN I-score was evaluated in nine patients during follow-up. The simultaneous reduction of IFN I-score and its components, CMAS and CAT scores was observed. Conclusion Skin involvement in refractory JDM is a challenging problem requiring the use of additional medications. Serum IFN I-score might be suggested as the promising biomarker of skin disease activity in JDM patients. Further investigations on patients with JDM and recurrent disease activity are needed, especially concerning biomarkers that determine the response to JAK inhibitors and treatment options for patients who don't respond to them.
Collapse
Affiliation(s)
- Rinat Raupov
- Saint-Petersburg State Pediatric Medical University, Saint Petersburg, Russia
- H.Turner National Medical Research Center for Children’s Orthopedics and Trauma Surgery, Saint Petersburg, Russia
| | - Evgeny Suspitsin
- Saint-Petersburg State Pediatric Medical University, Saint Petersburg, Russia
- N.N.Petrov Institute of Oncology, Saint Petersburg, Russia
| | | | - Mikhail Kostik
- Saint-Petersburg State Pediatric Medical University, Saint Petersburg, Russia
- Research Laboratory of Autoimmune and Autoinflammatory Diseases, World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russia
| |
Collapse
|
15
|
Stawicki MK, Abramowicz P, Sokolowska G, Wołejszo S, Grant WB, Konstantynowicz J. Can vitamin D be an adjuvant therapy for juvenile rheumatic diseases? Rheumatol Int 2023; 43:1993-2009. [PMID: 37566255 PMCID: PMC10495493 DOI: 10.1007/s00296-023-05411-5] [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: 07/14/2023] [Accepted: 07/27/2023] [Indexed: 08/12/2023]
Abstract
Vitamin D, known for its essential role in calcium and bone homeostasis, has multiple effects beyond the skeleton, including regulation of immunity and modulation of autoimmune processes. Several reports have shown suboptimal serum 25 hydroxyvitamin D [25(OH)D] levels in people with different inflammatory and autoimmune rheumatic conditions, and an association between 25(OH)D levels, disease activity and outcomes. Although most available data pertain to adults, insights often are extended to children. Juvenile rheumatic diseases (JRDs) are a significant health problem during growth because of their complex pathogenesis, chronic nature, multisystemic involvement, and long-term consequences. So far, there is no definitive or clear evidence to confirm the preventive or therapeutic effect of vitamin D supplementation in JRDs, because results from randomized controlled trials (RCTs) have produced inconsistent outcomes. This review aims to explore and discuss the potential role of vitamin D in treating selected JRDs. Medline/PubMed, EMBASE, and Scopus were comprehensively searched in June 2023 for any study on vitamin D supplementary role in treating the most common JRDs. We used the following keywords: "vitamin D" combined with the terms "juvenile idiopathic arthritis", "juvenile systemic scleroderma", "juvenile systemic lupus erythematosus", "juvenile inflammatory myopathies", "Behcet disease", "periodic fever syndromes" and "juvenile rheumatic diseases". Observational studies have found that serum 25(OH)D concentrations are lower in juvenile idiopathic arthritis, juvenile systemic lupus erythematosus, juvenile systemic scleroderma, Behcet disease and proinflammatory cytokine concentrations are higher. This suggests that vitamin D supplementation might be beneficial, however, current data are insufficient to confirm definitively the complementary role of vitamin D in the treatment of JRDs. Considering the high prevalence of vitamin D deficiency worldwide, children and adolescents should be encouraged to supplement vitamin D according to current recommendations. More interventional studies, especially well-designed RCTs, assessing the dose-response effect and adjuvant effect in specific diseases, are needed to determine the potential significance of vitamin D in JRDs treatment.
Collapse
Affiliation(s)
- Maciej K. Stawicki
- Department of Pediatrics, Rheumatology, Immunology, and Metabolic Bone Diseases, Medical University of Bialystok, University Children’s Clinical Hospital in Bialystok, Waszyngtona Street 17, 15274 Bialystok, Poland
| | - Paweł Abramowicz
- Department of Pediatrics, Rheumatology, Immunology, and Metabolic Bone Diseases, Medical University of Bialystok, University Children’s Clinical Hospital in Bialystok, Waszyngtona Street 17, 15274 Bialystok, Poland
| | | | - Sebastian Wołejszo
- Department of Pediatrics, Rheumatology, Immunology, and Metabolic Bone Diseases, Medical University of Bialystok, University Children’s Clinical Hospital in Bialystok, Waszyngtona Street 17, 15274 Bialystok, Poland
| | - William B. Grant
- Sunlight, Nutrition, and Health Research Center, San Francisco, CA USA
| | - Jerzy Konstantynowicz
- Department of Pediatrics, Rheumatology, Immunology, and Metabolic Bone Diseases, Medical University of Bialystok, University Children’s Clinical Hospital in Bialystok, Waszyngtona Street 17, 15274 Bialystok, Poland
| |
Collapse
|
16
|
Sherman MA, Pak K, Pinal-Fernandez I, Flegel WA, Targoff IN, Miller FW, Rider LG, Mammen AL, Childhood Myositis Heterogeneity Collaborative Study Group. Autoantibodies Recognizing Specificity Protein 4 Co-occur With Anti-Transcription Intermediary Factor 1 and Are Associated With Distinct Clinical Features and Immunogenetic Risk Factors in Juvenile Myositis. Arthritis Rheumatol 2023; 75:1668-1677. [PMID: 36996276 PMCID: PMC10524257 DOI: 10.1002/art.42512] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 03/17/2023] [Accepted: 03/28/2023] [Indexed: 04/01/2023]
Abstract
OBJECTIVE Autoantibodies recognizing specificity protein 4 (Sp4) were recently discovered in adults with idiopathic inflammatory myopathies (IIM). Anti-Sp4 autoantibodies co-occurred in patients with anti-transcription intermediary factor 1 (anti-TIF1) autoantibody-positive dermatomyositis (DM) and were associated with a reduced risk of cancer. In the present study, the prevalence and clinical features associated with anti-Sp4 autoantibodies in juvenile-onset IIM were investigated. METHODS Serum samples from 336 patients with juvenile myositis in a cross-sectional cohort and 91 healthy controls were screened for anti-Sp4 autoantibodies using enzyme-linked immunosorbent assay. Clinical characteristics, outcomes, and HLA alleles of those with and those without anti-Sp4 autoantibodies were compared. RESULTS Anti-Sp4 autoantibodies were present in 23 patients (7%) with juvenile myositis and were not present in any of the controls. Anti-Sp4 autoantibodies were found among each clinical myositis subgroup. The frequency of TIF1 autoantibody positivity was significantly higher among those with anti-Sp4 autoantibodies (21 [91%] versus 92 [30%], P < 0.001). In the anti-TIF1 autoantibody-positive subgroup, Raynaud's phenomenon (8 [38%] versus 2 [2%], P < 0.001) was more common and peak aspartate aminotransferase was significantly lower in those with anti-Sp4 autoantibodies. None of the patients with anti-Sp4 autoantibodies required a wheelchair. Among White patients, DQA1*04 and DRB1*08 were associated with anti-Sp4 autoantibodies. CONCLUSION Anti-Sp4 autoantibodies were found in patients with juvenile-onset IIM, predominantly those with coexisting anti-TIF1 autoantibodies. Patients with anti-Sp4 autoantibodies represent a phenotypic subset of anti-TIF1 autoantibody-positive myositis characterized by frequent Raynaud's phenomenon and less pronounced muscle involvement, similar to adults with these autoantibodies. Novel immunogenetic risk factors for White patients with IIM were identified among juveniles with anti-Sp4 autoantibodies.
Collapse
Affiliation(s)
- Matthew A. Sherman
- Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Katherine Pak
- Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Iago Pinal-Fernandez
- Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Willy A. Flegel
- Department of Transfusion Medicine, National Institutes of Health Clinical Center, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Ira N. Targoff
- Veteran’s Affairs Medical Center, University of Oklahoma Health Sciences Center, and Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Frederick W. Miller
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Lisa G. Rider
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Andrew L. Mammen
- Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | |
Collapse
|
17
|
Kumar A, Prajapati P, Singh G, Kumar D, Mishra V, Kim SC, Raorane CJ, Raj V, Kushwaha S. Salbutamol Attenuates Diabetic Skeletal Muscle Atrophy by Reducing Oxidative Stress, Myostatin/GDF-8, and Pro-Inflammatory Cytokines in Rats. Pharmaceutics 2023; 15:2101. [PMID: 37631314 PMCID: PMC10458056 DOI: 10.3390/pharmaceutics15082101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/02/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Type 2 diabetes is a metabolic disorder that leads to accelerated skeletal muscle atrophy. In this study, we aimed to evaluate the effect of salbutamol (SLB) on skeletal muscle atrophy in high-fat diet (HFD)/streptozotocin (STZ)-induced diabetic rats. Male Sprague Dawley rats were divided into four groups (n = 6): control, SLB, HFD/STZ, and HFD/STZ + SLB (6 mg/kg orally for four weeks). After the last dose of SLB, rats were assessed for muscle grip strength and muscle coordination (wire-hanging, rotarod, footprint, and actophotometer tests). Body composition was analyzed in live rats. After that, animals were sacrificed, and serum and gastrocnemius (GN) muscles were collected. Endpoints include myofibrillar protein content, muscle oxidative stress and antioxidants, serum pro-inflammatory cytokines (interleukin-1β, interleukin-2, and interleukin-6), serum muscle markers (myostatin, creatine kinase, and testosterone), histopathology, and muscle 1H NMR metabolomics. Findings showed that SLB treatment significantly improved muscle strength and muscle coordination, as well as increased lean muscle mass in diabetic rats. Increased pro-inflammatory cytokines and muscle markers (myostatin, creatine kinase) indicate muscle deterioration in diabetic rats, while SLB intervention restored the same. Also, Feret's diameter and cross-sectional area of GN muscle were increased by SLB treatment, indicating the amelioration in diabetic rat muscle. Results of muscle metabolomics exhibit that SLB treatment resulted in the restoration of perturbed metabolites, including histidine-to-tyrosine, phenylalanine-to-tyrosine, and glutamate-to-glutamine ratios and succinate, sarcosine, and 3-hydroxybutyrate (3HB) in diabetic rats. These metabolites showed a pertinent role in muscle inflammation and oxidative stress in diabetic rats. In conclusion, findings showed that salbutamol could be explored as an intervention in diabetic-associated skeletal muscle atrophy.
Collapse
Affiliation(s)
- Anand Kumar
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India; (A.K.); (P.P.); (V.M.)
| | - Priyanka Prajapati
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India; (A.K.); (P.P.); (V.M.)
| | - Gurvinder Singh
- Centre of Biomedical Research, SGPGIMS Campus, Lucknow 226014, India; (G.S.); (D.K.)
| | - Dinesh Kumar
- Centre of Biomedical Research, SGPGIMS Campus, Lucknow 226014, India; (G.S.); (D.K.)
| | - Vikas Mishra
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India; (A.K.); (P.P.); (V.M.)
| | - Seong-Cheol Kim
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | | | - Vinit Raj
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | - Sapana Kushwaha
- National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, New Transit Campus, Bijnor-Sisendi Road, Lucknow 226002, India
| |
Collapse
|
18
|
Papadopoulou C, Chew C, Wilkinson MGL, McCann L, Wedderburn LR. Juvenile idiopathic inflammatory myositis: an update on pathophysiology and clinical care. Nat Rev Rheumatol 2023; 19:343-362. [PMID: 37188756 PMCID: PMC10184643 DOI: 10.1038/s41584-023-00967-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2023] [Indexed: 05/17/2023]
Abstract
The childhood-onset or juvenile idiopathic inflammatory myopathies (JIIMs) are a heterogenous group of rare and serious autoimmune diseases of children and young people that predominantly affect the muscles and skin but can also involve other organs, including the lungs, gut, joints, heart and central nervous system. Different myositis-specific autoantibodies have been identified that are associated with different muscle biopsy features, as well as with different clinical characteristics, prognoses and treatment responses. Thus, myositis-specific autoantibodies can be used to subset JIIMs into sub-phenotypes; some of these sub-phenotypes parallel disease seen in adults, whereas others are distinct from adult-onset idiopathic inflammatory myopathies. Although treatments and management have much improved over the past decade, evidence is still lacking for many of the current treatments and few validated prognostic biomarkers are available with which to predict response to treatment, comorbidities (such as calcinosis) or outcome. Emerging data on the pathogenesis of the JIIMs are leading to proposals for new trials and tools for monitoring disease.
Collapse
Affiliation(s)
- Charalampia Papadopoulou
- Department of Paediatric Rheumatology, Great Ormond Street Hospital for Children NHS Foundation Trust (GOSH), London, UK
- Rare Diseases Theme NIHR Biomedical Research Centre at GOSH, London, UK
| | - Christine Chew
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Meredyth G Ll Wilkinson
- Rare Diseases Theme NIHR Biomedical Research Centre at GOSH, London, UK
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, UK
- Infection Immunity and Inflammation Research and Teaching Department, UCL GOS Institute of Child Health, London, UK
| | - Liza McCann
- Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust, Liverpool, UK
| | - Lucy R Wedderburn
- Department of Paediatric Rheumatology, Great Ormond Street Hospital for Children NHS Foundation Trust (GOSH), London, UK.
- Rare Diseases Theme NIHR Biomedical Research Centre at GOSH, London, UK.
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, UK.
- Infection Immunity and Inflammation Research and Teaching Department, UCL GOS Institute of Child Health, London, UK.
| |
Collapse
|
19
|
Fujiwara M, Wakiguchi H, Okazaki F, Korenaga Y, Uchida M, Sato R, Omoto M, Kanda T, Hasegawa S. Magnetic resonance imaging and pathological findings of fasciitis in anti-PM/Scl antibody-positive juvenile myositis. Int J Rheum Dis 2023; 26:577-580. [PMID: 36504181 DOI: 10.1111/1756-185x.14519] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 12/14/2022]
Affiliation(s)
- Mayu Fujiwara
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Hiroyuki Wakiguchi
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Fumiko Okazaki
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Yuno Korenaga
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Masashi Uchida
- Division of Pediatrics, JCHO Tokuyama Central Hospital, Shunan, Japan
| | - Ryota Sato
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Masatoshi Omoto
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Takashi Kanda
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Shunji Hasegawa
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Ube, Japan
| |
Collapse
|
20
|
Sherman MA, Kim H, Banschbach K, Brown A, Gewanter HL, Lang B, Perron M, Robinson AB, Spitznagle J, Stingl C, Syverson G, Tory HO, Spencer CH, Tarvin SE. Treatment escalation patterns to start biologics in refractory moderate juvenile dermatomyositis among members of the Childhood Arthritis and Rheumatology Research Alliance. Pediatr Rheumatol Online J 2023; 21:3. [PMID: 36609397 PMCID: PMC9825021 DOI: 10.1186/s12969-022-00785-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 12/26/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Despite new and better treatments for juvenile dermatomyositis (JDM), not all patients with moderate severity disease respond adequately to first-line therapy. Those with refractory disease remain at higher risk for disease and glucocorticoid-related complications. Biologic disease-modifying antirheumatic drugs (DMARDs) have become part of the arsenal of treatments for JDM. However, prospective comparative studies of commonly used biologics are lacking. METHODS The Childhood Arthritis and Rheumatology Research Alliance (CARRA) JDM biologics workgroup met in 2019 and produced a survey assessing current treatment escalation practices for JDM, including preferences regarding use of biologic treatments. The cases and questions were developed using a consensus framework, requiring 80% agreement for consensus. The survey was completed online in 2020 by CARRA members interested in JDM. Survey results were analyzed among all respondents and according to years of experience. Chi-square or Fisher's exact test was used to compare the distribution of responses to each survey question. RESULTS One hundred twenty-one CARRA members responded to the survey (denominators vary for each question). Of the respondents, 88% were pediatric rheumatologists, 85% practiced in the United States, and 43% had over 10 years of experience. For a patient with moderately severe JDM refractory to methotrexate, glucocorticoids, and IVIG, approximately 80% of respondents indicated that they would initiate a biologic after failing 1-2 non-biologic DMARDs. Trials of methotrexate and mycophenolate were considered necessary by 96% and 60% of respondents, respectively, before initiating a biologic. By weighed average, rituximab was the preferred biologic over abatacept, tocilizumab, and infliximab. Over 50% of respondents would start a biologic by 4 months from diagnosis for patients with refractory moderately severe JDM. There were no notable differences in treatment practices between respondents by years of experience. CONCLUSION Most respondents favored starting a biologic earlier in disease course after trialing up to two conventional DMARDs, specifically including methotrexate. There was a clear preference for rituximab. However, there remains a dearth of prospective data comparing biologics in refractory JDM. These findings underscore the need for biologic consensus treatment plans (CTPs) for refractory JDM, which will ultimately facilitate comparative effectiveness studies and inform treatment practices.
Collapse
Affiliation(s)
- Matthew A. Sherman
- grid.94365.3d0000 0001 2297 5165Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 9000 Rockville Pike 50 South Drive Building 50, Room 1142, 20892 Bethesda, MD USA ,grid.239560.b0000 0004 0482 1586Division of Rheumatology, Children’s National Hospital, Washington, DC USA
| | - Hanna Kim
- grid.420086.80000 0001 2237 2479Juvenile Myositis Pathogenesis and Therapeutics Unit, National Institute of Arthritis Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD USA
| | - Katelyn Banschbach
- grid.240741.40000 0000 9026 4165Division of Rheumatology, Department of Pediatrics, University of Washington/Seattle Children’s Hospital, Seattle, WA USA
| | - Amanda Brown
- grid.241054.60000 0004 4687 1637University of Arkansas for Medical Sciences and Arkansas Children’s Hospital, Little Rock, AR USA
| | - Harry L. Gewanter
- grid.414220.1Children’s Hospital of Richmond at VCU, Richmond, VA USA
| | - Bianca Lang
- grid.55602.340000 0004 1936 8200IWK Health, Dalhousie University, Halifax, NS Canada
| | - Megan Perron
- grid.413957.d0000 0001 0690 7621Department of Pediatric Rheumatology, Children’s Hospital Colorado, Aurora, CO USA
| | - Angela Byun Robinson
- grid.239578.20000 0001 0675 4725Pediatric Rheumatology, Cleveland Clinic Children’s Hospital, Cleveland, OH USA
| | - Jacob Spitznagle
- grid.240741.40000 0000 9026 4165Division of Rheumatology, Department of Pediatrics, University of Washington/Seattle Children’s Hospital, Seattle, WA USA
| | - Cory Stingl
- grid.416230.20000 0004 0406 3236Department of Pediatrics, Spectrum Health, Grand Rapids, MI USA
| | - Grant Syverson
- grid.490404.d0000 0004 0425 6409Sanford Health, Fargo, ND USA
| | - Heather O. Tory
- grid.63054.340000 0001 0860 4915Connecticut Children’s Medical Center, Hartford, CT, USA and University of Connecticut School of Medicine, Farmington, Farmington, CT USA
| | - Charles H. Spencer
- grid.410721.10000 0004 1937 0407University of Mississippi Medical Center, Batson Children’s Hospital, Jackson, MS USA
| | - Stacey E. Tarvin
- grid.257413.60000 0001 2287 3919Division of Rheumatology, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN USA
| |
Collapse
|
21
|
Muacevic A, Adler JR, Chaudhari J, Panchal V, Shah A. An Unusual Presentation of Juvenile Polymyositis in an Adolescent Girl. Cureus 2023; 15:e33249. [PMID: 36741666 PMCID: PMC9890880 DOI: 10.7759/cureus.33249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2023] [Indexed: 01/03/2023] Open
Abstract
Juvenile idiopathic inflammatory myopathies (JIIMs) are a group of diverse, systemic autoimmune diseases that manifest in childhood and are characterized by weakness and chronic inflammation of skeletal muscles. One of the relatively rare variants of JIIMs is juvenile polymyositis (JPM). JPM patients present with proximal and distal muscle weakness, gait instability with falls, muscle pain and tenderness, and high levels of creatine kinase (CK) during adolescence. There are currently few people being diagnosed with JPM, which raises the question of whether or not it is a distinct disease. We discuss the case of a 13-year-old girl who presented to the hospital with generalized body swelling and difficulty swallowing solid food. She also had drooling of saliva during the presentation and a history of difficulty climbing up and down the stairs for three months. Her extensive laboratory workup showed a positive antinuclear antibody (ANA) test and increased muscle enzyme. A muscle biopsy was ordered, and she was diagnosed with JPM. Such a unique presentation has rarely been reported in the pediatric literature. This case report outlines an unusual JPM presentation that could help clinicians identify the condition and start treatment as soon as possible to minimize complications.
Collapse
|
22
|
Clinical and autoantibody phenotypes of juvenile dermatomyositis. Reumatologia 2022; 60:281-291. [PMID: 36186835 PMCID: PMC9494788 DOI: 10.5114/reum.2022.119045] [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: 05/12/2022] [Accepted: 07/25/2022] [Indexed: 11/17/2022] Open
Abstract
Juvenile dermatomyositis (JDM) is a heterogeneous autoimmune inflammatory myositis with symmetrical proximal muscle weakness and a characteristic rash. Juvenile dermatomyositis is characterized by variable presentation and phenotypes. Detection of myositis autoantibodies is useful in improving JDM diagnosis and predicting the prognosis. In this literature review based on case series we analyze clinical and autoantibody phenotypes of JDM in four patients who were hospitalized in one regional center in Ukraine during the last 3 years and three of them presented in the time of the COVID-19 pandemic. The reviewed literature showed the last updates for the JDM diagnosis and the role of myositis autoantibodies in the prediction of disease course, systemic involvement, and malignancy risk. The presence of anti-synthetase syndrome in all presented patients, mainly due to anti-PL-7 autoantibodies, encourages further study with more patients and with detection of other myositis-specific autoantibodies to identify or refute certain regional features.
Collapse
|
23
|
Radziszewska A, Moulder Z, Jury EC, Ciurtin C. CD8 + T Cell Phenotype and Function in Childhood and Adult-Onset Connective Tissue Disease. Int J Mol Sci 2022; 23:11431. [PMID: 36232733 PMCID: PMC9569696 DOI: 10.3390/ijms231911431] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/14/2022] [Accepted: 09/19/2022] [Indexed: 11/21/2022] Open
Abstract
CD8+ T cells are cytotoxic lymphocytes that destroy pathogen infected and malignant cells through release of cytolytic molecules and proinflammatory cytokines. Although the role of CD8+ T cells in connective tissue diseases (CTDs) has not been explored as thoroughly as that of other immune cells, research focusing on this key component of the immune system has recently gained momentum. Aberrations in cytotoxic cell function may have implications in triggering autoimmunity and may promote tissue damage leading to exacerbation of disease. In this comprehensive review of current literature, we examine the role of CD8+ T cells in systemic lupus erythematosus, Sjögren's syndrome, systemic sclerosis, polymyositis, and dermatomyositis with specific focus on comparing what is known about CD8+ T cell peripheral blood phenotypes, CD8+ T cell function, and CD8+ T cell organ-specific profiles in adult and juvenile forms of these disorders. Although, the precise role of CD8+ T cells in the initiation of autoimmunity and disease progression remains to be elucidated, increasing evidence indicates that CD8+ T cells are emerging as an attractive target for therapy in CTDs.
Collapse
Affiliation(s)
- Anna Radziszewska
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH), Great Ormond Street Hospital (GOSH), London WC1E 6JF, UK
- Centre for Rheumatology Research, Division of Medicine, University College London, London WC1E 6JF, UK
| | - Zachary Moulder
- University College London Medical School, University College London, London WC1E 6DE, UK
| | - Elizabeth C. Jury
- Centre for Rheumatology Research, Division of Medicine, University College London, London WC1E 6JF, UK
| | - Coziana Ciurtin
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH), Great Ormond Street Hospital (GOSH), London WC1E 6JF, UK
- Centre for Rheumatology Research, Division of Medicine, University College London, London WC1E 6JF, UK
| |
Collapse
|
24
|
Sherman MA, Graf R, Sabbagh SE, Galindo-Feria AS, Pinal-Fernandez I, Pak K, Kishi T, Flegel WA, Targoff IN, Miller FW, Lundberg IE, Rider LG, Mammen AL. Anti-FHL1 autoantibodies in juvenile myositis are associated with anti-Ro52 autoantibodies but not with severe disease features. Rheumatology (Oxford) 2022; 62:SI226-SI234. [PMID: 35961028 PMCID: PMC9949705 DOI: 10.1093/rheumatology/keac428] [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: 05/04/2022] [Revised: 07/17/2022] [Accepted: 07/17/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES Four-and-a-half LIM domains 1 (FHL1) is a muscle-specific protein. Autoantibodies against FHL1 were recently discovered in adults with idiopathic inflammatory myopathies (IIMs) and were found to be associated with clinical features and outcomes indicative of increased disease severity. Anti-FHL1 autoantibodies have not been described in children. Here, the prevalence and clinical features associated with anti-FHL1 autoantibodies were examined in a large North American cohort of juvenile patients with IIM. METHODS Sera from 338 juvenile IIM patients and 91 juvenile healthy controls were screened for anti-FHL1 autoantibodies by ELISA. Clinical characteristics and HLA alleles of those with and without anti-FHL1 autoantibodies were compared among those with juvenile IIM. RESULTS Anti-FHL1 autoantibodies were present in 10.9% of juvenile IIM patients and 1.1% of controls. The frequency of anti-FHL1 autoantibodies among clinical and serologic subgroups did not differ. A higher percentage of Asian patients had anti-FHL1 autoantibodies (11% vs 0.7%; P = 0.002). Myositis-associated autoantibodies (MAAs) [odds ratio (OR) 2.09 (CI 1.03, 4.32)], anti-Ro52 autoantibodies specifically [OR 4.17 (CI 1.83, 9.37)] and V-sign rash [OR 2.59 (CI 1.22, 5.40)] were associated with anti-FHL1 autoantibodies. There were no differences in other features or markers of disease severity. No HLA associations with anti-FHL1 autoantibodies in Caucasian myositis patients were identified. CONCLUSION Anti-FHL1 autoantibodies are present in ∼11% of juvenile IIM patients and commonly co-occur with MAAs, including anti-Ro52 autoantibodies. In contrast to adult IIM, anti-FHL1 autoantibodies in juvenile myositis are associated with V-sign rash but not with other distinctive clinical features or worse outcomes.
Collapse
Affiliation(s)
- Matthew A Sherman
- Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | | | | | - Angeles S Galindo-Feria
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet,Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | | | - Katherine Pak
- Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Takayuki Kishi
- Environmental Autoimmunity Group, National Institute of Environmental Health Sciences
| | - Willy A Flegel
- Department of Transfusion Medicine, NIH Clinical Center, National Institutes of Health, Bethesda, MD
| | - Ira N Targoff
- Veteran's Affairs Medical Center, University of Oklahoma Health Sciences Center, and Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Frederick W Miller
- Environmental Autoimmunity Group, National Institute of Environmental Health Sciences
| | - Ingrid E Lundberg
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet,Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | | | - Andrew L Mammen
- Correspondence to: Andrew L. Mammen, Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Expression, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 50 South Drive, Room 1141, Building 50, MSC 8024, Bethesda, MD 20892, USA. E-mail:
| | | |
Collapse
|
25
|
Neely J, Ardalan K, Huber A, Kim S. Baseline characteristics of children with juvenile dermatomyositis enrolled in the first year of the new Childhood Arthritis and Rheumatology Research Alliance registry. Pediatr Rheumatol Online J 2022; 20:50. [PMID: 35854378 PMCID: PMC9295519 DOI: 10.1186/s12969-022-00709-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 07/02/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND To report baseline characteristics, patient reported outcomes and treatment of children with Juvenile Dermatomyositis (JDM) in the Childhood Arthritis and Rheumatology Research Alliance (CARRA) Registry. METHODS Children newly diagnosed with JDM were enrolled in the CARRA Registry from 41 pediatric rheumatology centers. Baseline patient demographics, disease characteristics, assessments, patient reported outcome and treatments were recorded. RESULTS In the first year, 119 JDM participants were enrolled. Most were female (63.4%), and white (72.3%) with a median diagnosis age 8.0 years (IQR 4.0-11.5), and median age of disease onset 7.0 years (IQR 3.5-7.5). They had characteristic rashes (92.4%), elevated muscle enzymes (83.2%), physician global score 4.0 (IQR 2.5-5.0) and manual muscle testing score 63.5 (IQR 51.0-75.0). Calcinosis (3.4%) and interstitial lung disease (< 1%) were uncommon. Myositis specific antibodies were measured and reported in nearly half of participants enrolled where anti-MJ followed by Anti-p155/140 were most common (11/49 and 7/53 respectively). Childhood Health Assessment Questionnaire (CHAQ) results showed mild-moderate disability (median 0.750, IQR 0.030-1.875), as did patient/parent global assessments of disease activity (median 3, patient IQR: 1.75-5.25; parent IQR: 1-7). Patient Reported Outcomes Measurement Information System (PROMIS®) Pediatric Global Health 7 scores, Pain Interference, Physical Function scores for Mobility, and Upper Extremity Function were commonly worse than 95% of the general pediatric population. CONCLUSIONS In its inaugural year, 119 JDM patients were successfully enrolled in participapte in the New CARRA Registy. This registry will provide the necessary foundation to advance clinical research to improve outcomes using traditional measures and patient reported outcomes. With the CARRA biorepository, this infrastructure will enable future translational research. Together, these efforts may aid in future clinical trials, including comparative effectiveness trials.
Collapse
Affiliation(s)
- Jessica Neely
- grid.413077.60000 0004 0434 9023University of California San Francisco Medical Center, 550 16th Street, San Francisco, CA 94158 USA
| | - Kaveh Ardalan
- grid.189509.c0000000100241216Duke University Medical Center, 2301 Erwin Rd, Durham, NC 27705 USA
| | - Adam Huber
- grid.414870.e0000 0001 0351 6983Division of Pediatric Rheumatology, IWK Health Centre and Dalhousie University, PO Box 9700, 5850-5980 University Ave, Halifax, Nova Scotia Canada
| | - Susan Kim
- University of California San Francisco Medical Center, 550 16th Street, San Francisco, CA, 94158, USA.
| | | |
Collapse
|
26
|
Karasawa R, Yudoh K, Sato T, Tanaka M, Tamaki M, Sabbagh SE, O’Hanlon TP, Noroozi-Farhadi P, Targoff IN, Flegel WA, Mammen AL, Miller FW, Hicar MD, Rider LG, Jarvis JN. Association of anti-HSC70 autoantibodies with cutaneous ulceration and severe disease in juvenile dermatomyositis. Rheumatology (Oxford) 2022; 61:2969-2977. [PMID: 34791087 PMCID: PMC9258543 DOI: 10.1093/rheumatology/keab846] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 11/06/2021] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES JDM is an inflammatory myopathy characterized by prominent vasculopathy. AECAs are frequently detected in inflammatory and autoimmune diseases. We sought to determine whether AECAs correlate with clinical features of JDM, and thus serve as biomarkers to guide therapy or predict outcome. METHODS Plasma samples from 63 patients with JDM, 49 patients with polyarticular JIA and 40 juvenile healthy controls were used to detect anti-heat shock cognate 71 kDa protein (HSC70) autoantibodies, a newly identified AECA, in ELISA assays. Clinical features were compared between JDM patients with and without anti-HSC70 autoantibodies. RESULTS Anti-HSC70 autoantibodies were detected in 35% of patients with JDM, in 0% of patients with JIA (P < 0.0001) and in 0% of healthy donors (P < 0.0001). Both the presence of cutaneous ulcers (59% vs 17%, P < 0.002) and the use of wheelchairs and/or assistive devices (64% vs 27%, P < 0.007) were strongly associated with anti-HSC70 autoantibodies in JDM. High scores on the severity of myositis damage measures at the time of measurement of anti-HSC70 autoantibodies and an increased number of hospitalizations were also associated with anti-HSC70 autoantibodies. Intravenous immunoglobulin therapy was used more often in anti-HSC70 autoantibody-positive patients. CONCLUSION Anti-HCS70 autoantibodies are detected frequently in children with JDM and are novel myositis-associated autoantibodies correlating with disease severity.
Collapse
Affiliation(s)
- Rie Karasawa
- Department of Frontier Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Kazuo Yudoh
- Department of Frontier Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Toshiko Sato
- Department of Frontier Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Megumi Tanaka
- Department of Frontier Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Mayumi Tamaki
- Department of Frontier Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Sara E Sabbagh
- Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health (NIH), Bethesda, MD
- Division of Rheumatology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
| | - Terrance P O’Hanlon
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health (NIH), Bethesda, MD
| | - Payam Noroozi-Farhadi
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health (NIH), Bethesda, MD
| | - Ira N Targoff
- Oklahoma City VA Health Care System, University of Oklahoma Health Sciences Center, and Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Willy A Flegel
- Department of Transfusion Medicine, NIH Clinical Center, National Institutes of Health (NIH), Bethesda, MD
| | - Andrew L Mammen
- Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health (NIH), Bethesda, MD
| | - Frederick W Miller
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health (NIH), Bethesda, MD
| | - Mark D Hicar
- Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences
| | - Lisa G Rider
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health (NIH), Bethesda, MD
| | - James N Jarvis
- Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences
- Genetics, Genomics, & Bioinformatics Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
27
|
Mondal S, Barman P, Vignesh P. Cardiovascular Abnormalities in Juvenile Dermatomyositis: A Scoping Review for the Clinical Rheumatologists. Front Med (Lausanne) 2022; 9:827539. [PMID: 35814777 PMCID: PMC9263083 DOI: 10.3389/fmed.2022.827539] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 05/09/2022] [Indexed: 11/17/2022] Open
Abstract
Juvenile dermatomyositis (JDM) is a common form of inflammatory myositis in children. Vasculopathy and endothelial dysfunction play significant roles in the pathogenesis of JDM. Cardiac involvement in JDM is often underestimated, and it may be a potential indicator of poor prognosis. Cardiac dysfunction in JDM can occur both in the acute and chronic stages of the disease. Amongst the acute complications, acute congestive heart failure (CHF), myocarditis, arrhythmia, and complete heart block are common. However, these remain unrecognized due to a lack of overt clinical manifestations. Increased rates of cardiovascular abnormalities have been noted with anti-SRP and anti-Jo 1 auto-antibody positivity. Long-term follow-up studies in JDM have shown an increased prevalence of hypertension, atherosclerosis, coronary artery disease, and metabolic syndrome in adolescence and adulthood. Monitoring of body-mass index, blood pressure, and laboratory evaluation of fasting glucose and lipid profile may help in identifying metabolic syndrome in children with JDM. Steroid-sparing agents, daily exercise, and a healthy diet may reduce such long-term cardiac morbidities. Current use of multimodality imaging such as stress-echocardiography, contrast-enhanced echocardiography, cardiac magnetic resonance imaging, and positron emission tomography has increased the diagnostic yield of subclinical heart disease during acute and chronic stages of JDM. This review elaborates on different aspects of cardiac dysfunction in JDM. It also emphasizes the importance of cardiac screening in long-term follow-up of children with JDM.
Collapse
Affiliation(s)
| | | | - Pandiarajan Vignesh
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
28
|
Stingl C, Dvergsten JA, Eng SWM, Yeung RSM, Fritzler MJ, Mason T, Crowson C, Voora D, Reed AM. Gene Expression Profiles of Treatment Response and Non-Response in Children With Juvenile Dermatomyositis. ACR Open Rheumatol 2022; 4:671-681. [PMID: 35616642 PMCID: PMC9374052 DOI: 10.1002/acr2.11445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 01/26/2022] [Accepted: 02/01/2022] [Indexed: 12/26/2022] Open
Abstract
Objective The study objective was to identify differences in gene expression between treatment responders (TRs) and treatment non‐responders (TNRs) diagnosed with juvenile dermatomyositis (JDM). Methods Gene expression analyses were performed using whole blood messenger RNA sequencing in patients with JDM (n = 17) and healthy controls (HCs; n = 10). Four analyses were performed (A1‐4) comparing differential gene expression and pathways analysis exploiting the timing of sample acquisition and the treatments received to perform these comparative analyses. Analyses were done at diagnosis and follow‐up, which averaged 7 months later in the cohort. Results At diagnosis, the expression of 10 genes differed between TRs and TNRs. Hallmark and canonical pathway analysis revealed 11 and 60 pathways enriched in TRs and 3 and 21 pathways enriched in TNRs, respectively. Pathway enrichment at diagnosis in TRs was strongest in pathways involved in metabolism, complement activation, and cell signaling as mediated by IL‐8, p38/microtubule associated protein kinases (MAPK)/extracellular signal‐regulated kinases (ERK), Phosphatidylinositol 3 Kinase Gamma (PI3Kγ), and the B cell receptor. Follow‐up hallmark and canonical pathway analysis showed that 2 and 14 pathways were enriched in TRs, whereas 24 and 123 pathways were enriched in treatment TNRs, respectively. Prior treatment with glucocorticoids significantly altered expression of 13 genes in the analysis of subjects at diagnosis with JDM as compared with HCs. Conclusion Numerous genes and pathways differ between TRs and TNRs at diagnosis and follow‐up. Prior treatment with glucocorticoids prior to specimen acquisition had a small effect on the performed analyses.
Collapse
Affiliation(s)
| | | | - Simon W M Eng
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Rae S M Yeung
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
29
|
Gupta D, Goldberg L, Dickinson A, Hughes M, Anand V, Stokke J, Corden MH. An 8-Year-Old Boy With Prolonged Fever and Subcutaneous Nodules. Pediatrics 2022; 149:186710. [PMID: 35490281 DOI: 10.1542/peds.2021-052974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/16/2021] [Indexed: 11/24/2022] Open
Abstract
An 8-year-old boy with asthma presented with prolonged fever, malaise, extremity weakness, polyarthralgias, malar rash, and subcutaneous nodules. Physical examination was remarkable for a faint malar rash, flesh-colored papules on the dorsal aspect of the fingers, arthritis of multiple joints in the hands, and subcutaneous nodules. The nodules were firm, nontender, and distributed over multiple extremities and the trunk. The patient was admitted to expedite workup. Initial laboratory test results revealed leukopenia, mild elevation of the aminotransferases, an elevated erythrocyte sedimentation rate, and normal level of creatine kinase. His echocardiogram was normal. Infectious disease studies were negative. Additional examination revealed dilated capillaries in his nail beds and bilateral hip weakness. MRI of his extremities was negative for myositis or calcification of the nodules. We obtained a biopsy of the subcutaneous nodules, and because the patient remained afebrile during the hospitalization, we discharged him from the hospital with outpatient follow-up. Our expert panel reviews the course of the patient's evaluation and investigation, as well as the implications of his diagnosis based on the tissue pathology from the nodule biopsy.
Collapse
Affiliation(s)
| | | | | | - Meagan Hughes
- Dermatology.,Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Vikram Anand
- Infectious Diseases.,Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Jamie Stokke
- Cancer and Blood Disease Institute.,Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Mark H Corden
- Hospital Medicine, Department of Pediatrics, Children's Hospital Los Angeles.,Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
30
|
Environmental factors associated with juvenile idiopathic inflammatory myopathy clinical and serologic phenotypes. Pediatr Rheumatol Online J 2022; 20:28. [PMID: 35414090 PMCID: PMC9004071 DOI: 10.1186/s12969-022-00684-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/28/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Environmental exposures have been associated with the juvenile idiopathic inflammatory myopathies (JIIM). We undertook a questionnaire-based study to evaluate patient-reported exposures as possible risk factors for JIIM. FINDINGS One-hundred-seven patients with JIIM were enrolled in a myositis natural history protocol and completed environmental questionnaires. Frequencies of exposures in clinical and myositis-specific autoantibody (MSA) groups were examined. Patients with juvenile dermatomyositis (JDM) and juvenile connective tissue myositis (JCTM) more frequently received an immunization within 1 year of diagnosis compared to juvenile polymyositis (57.5 and 71.4% vs 0.0%, p ≤ 0.017). JCTM patients were more often underweight at diagnosis relative to JDM patients (42.9% vs 7.0%, p = 0.002). MSA-negative patients more frequently had gastroenteritis within a year of diagnosis compared to patients with anti-MDA5 autoantibodies (28.6% vs 0.0%, p = 0.032). Heavy exercise was more frequent in MSA-negative and anti-MDA5 groups compared to the anti-TIF-1 autoantibody group (42.9 and 35.3% vs. 9.0%, p ≤ 0.047). Medications received within 1 year of diagnosis were more frequent in MSA-negative patients relative to those with anti-MDA5 autoantibodies (92.9% vs. 52.8% p = 0.045). Being breastfed > 6 months was more frequent in MSA-negative patients (88.9%) compared to anti-TIF-1 and anti-MDA5 autoantibody groups (41.2 and 28.6%, p ≤ 0.036). CONCLUSIONS Certain environmental exposures prior to diagnosis differed among clinical and serologic subgroups of JIIM, suggesting additional exposures to be explored as possible risk factors for JIIM phenotypes.
Collapse
|
31
|
Pinal-Fernandez I, Pak K, Gil-Vila A, Baucells A, Plotz B, Casal-Dominguez M, Derfoul A, Martinez MA, Selva-O’Callaghan A, Sabbagh S, Casciola-Rosen L, Albayda J, Paik J, Tiniakou E, Danoff SK, Lloyd TE, Miller FW, Rider LG, Christopher-Stine L, Mammen AL. Anti-Cortactin Autoantibodies Are Associated With Key Clinical Features in Adult Myositis But Are Rarely Present in Juvenile Myositis. Arthritis Rheumatol 2022; 74:358-364. [PMID: 34313394 PMCID: PMC8792092 DOI: 10.1002/art.41931] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 06/15/2021] [Accepted: 06/22/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To define the prevalence and clinical phenotype of anti-cortactin autoantibodies in adult and juvenile myositis. METHODS In this longitudinal cohort study, anti-cortactin autoantibody titers were assessed by enzyme-linked immunosorbent assay in 670 adult myositis patients and 343 juvenile myositis patients as well as in 202 adult healthy controls and 90 juvenile healthy controls. The prevalence of anti-cortactin autoantibodies was compared among groups. Clinical features of patients with and those without anti-cortactin autoantibodies were also compared. RESULTS Anti-cortactin autoantibodies were more common in adult dermatomyositis (DM) patients (15%; P = 0.005), particularly those with coexisting anti-Mi-2 autoantibodies (24%; P = 0.03) or anti-NXP-2 autoantibodies (23%; P = 0.04). In adult myositis, anti-cortactin was associated with DM skin involvement (62% of patients with anti-cortactin versus 38% of patients without anti-cortactin; P = 0.03), dysphagia (36% versus 17%; P = 0.02) and coexisting anti-Ro 52 autoantibodies (47% versus 26%; P = 0.001) or anti-NT5c1a autoantibodies (59% versus 33%; P = 0.001). Moreover, the titers of anti-cortactin antibodies were higher in patients with interstitial lung disease (0.15 versus 0.12 arbitrary units; P = 0.03). The prevalence of anti-cortactin autoantibodies was not different in juvenile myositis patients (2%) or in any juvenile myositis subgroup compared to juvenile healthy controls (4%). Nonetheless, juvenile myositis patients with these autoantibodies had a higher prevalence of "mechanic's hands" (25% versus 7%; P = 0.03), a higher number of hospitalizations (2.9 versus 1.3; P = 0.04), and lower peak creatine kinase values (368 versus 818 IU/liter; P = 0.02) than those without anti-cortactin. CONCLUSION The prevalence of anti-cortactin autoantibodies is increased in adult DM patients with coexisting anti-Mi-2 or anti-NXP-2 autoantibodies. In adults, anti-cortactin autoantibodies are associated with dysphagia and interstitial lung disease.
Collapse
Affiliation(s)
- Iago Pinal-Fernandez
- Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Faculty of Health Sciences, and Faculty of Computer Science, Multimedia and Telecommunications, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Katherine Pak
- Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland
| | - Albert Gil-Vila
- Vall d’Hebron Hospital, Barcelona, Spain.,Autonomous University of Barcelona, Barcelona, Spain
| | | | - Benjamin Plotz
- Division of Rheumatology, New York University Langone Health, New York, NY
| | - Maria Casal-Dominguez
- Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Assia Derfoul
- Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland
| | | | | | - Sara Sabbagh
- Division of Rheumatology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Livia Casciola-Rosen
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jemima Albayda
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Julie Paik
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Eleni Tiniakou
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sonye K. Danoff
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Thomas E. Lloyd
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Frederick W. Miller
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, MD
| | - Lisa G. Rider
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, MD
| | - Lisa Christopher-Stine
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Andrew L. Mammen
- Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | | |
Collapse
|
32
|
Loarce-Martos J, Larena C, Blázquez MÁ, Joven BE, Carreira PE, Martínez-Barrio J, Monteagudo I, López-Longo FJ, Ruiz L, López-Robledillo JC, Almodóvar R, Llorente I, Tomero E, García-de la Peña P, Moruno H, Pérez A, Cobo-Ibáñez T, Lojo Oliveira L, Barbadillo MC, García-De Yébenes MJ, Nuño-Nuño L. Clinical Characteristics of Juvenile Idiopathic Inflammatory Myopathy and Comparison With Adult Patients: Analysis From a Multicentric Cohort in Spain. J Clin Rheumatol 2022; 28:e195-e202. [PMID: 33492027 DOI: 10.1097/rhu.0000000000001696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
METHODS This study reviewed the medical records of patients from the REMICAM cohort, a multicentric longitudinal study carried out in patients with IIM, followed up between 1980 and 2014 in 12 hospitals in Madrid, Spain. Patients with definite or probable JPM, JDM, adult DM, and adult PM according to the modified Bohan and Peter criteria were selected. We compared the characteristics between JDM and JPM, and between JIIM and adult IIM. RESULTS Eighty-six juvenile patients (75 JDMs and 11 JPMs) and 283 adult patients (133 DMs and 150 PMs) were included. Compared with patients with JDM, patients with JPM were older at diagnosis, had more fever and arthritis, and were less frequently treated with disease-modifying antirheumatic drugs (these differences were not statistically significant). Compared with patients with adult DM, those with JDM presented more frequently with calcinosis (33.8% vs 6.9%, p < 0.0001) and had less severe infections (4.3% vs 23.4%, p < 0.0001), malignancies (1.3% vs 25.6%, p < 0.0001), and mortality (3.5% vs 33%, p < 0.0001). Patients with JDM were treated less frequently with azathioprine (10.8% vs 44.7%, p < 0.0001). CONCLUSIONS Our findings confirm that JIIMs are a heterogeneous group of diseases with relevant differences compared with adult IIMs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Ana Pérez
- Hospital Universitario Príncipe de Asturias
| | | | | | | | | | | |
Collapse
|
33
|
Morphological Characteristics of Idiopathic Inflammatory Myopathies in Juvenile Patients. Cells 2021; 11:cells11010109. [PMID: 35011672 PMCID: PMC8750180 DOI: 10.3390/cells11010109] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/20/2021] [Accepted: 12/25/2021] [Indexed: 12/16/2022] Open
Abstract
Background: In juvenile idiopathic inflammatory myopathies (IIMs), morphological characteristic features of distinct subgroups are not well defined. New treatment strategies require a precise diagnosis of the subgroups in IIM, and, therefore, knowledge about the pathomorphology of juvenile IIMs is warranted. Methods: Muscle biopsies from 15 patients (median age 8 (range 3–17) years, 73% female) with IIM and seven controls were analyzed by standard methods, immunohistochemistry, and transmission electron microscopy (TEM). Detailed clinical and laboratory data were accessed retrospectively. Results: Proximal muscle weakness and skin symptoms were the main clinical symptoms. Dermatomyositis (DM) was diagnosed in 9/15, antisynthetase syndrome (ASyS) in 4/15, and overlap myositis (OM) in 2/15. Analysis of skeletal muscle tissues showed inflammatory cells and diffuse upregulation of MHC class I in all subtypes. Morphological key findings were COX-deficient fibers as a striking pathology in DM and perimysial alkaline phosphatase positivity in anti-Jo-1-ASyS. Vascular staining of the type 1 IFN-surrogate marker, MxA, correlated with endothelial tubuloreticular inclusions in both groups. None of these specific morphological findings were present in anti-PL7-ASyS or OM patients. Conclusions: Morphological characteristics discriminate IIM subtypes in juvenile patients, emphasizing differences in aetiopathogenesis and supporting the notion of individual and targeted therapeutic strategies.
Collapse
|
34
|
伊 文, 魏 翠, 吴 晔, 包 新, 熊 晖, 常 杏. [Long-term rituximab treatment of refractory idiopathic inflammatory myopathy: A report of 3 cases]. BEIJING DA XUE XUE BAO. YI XUE BAN = JOURNAL OF PEKING UNIVERSITY. HEALTH SCIENCES 2021; 53:1191-1195. [PMID: 34916704 PMCID: PMC8695162 DOI: 10.19723/j.issn.1671-167x.2021.06.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Indexed: 06/14/2023]
Abstract
Idiopathic inflammatory myopathies are a group of rare but serious diseases. The treatment of refractory idiopathic inflammatory myopathy is always challenging, especially in children. Three cases of refractory idiopathic inflammatory myopathy treated by rituximab were reported and discussed with the review of relevant literature. All were female with on-set age of 8 years and 6 months, 11 years and 7 months, 4 years and 2 months old, respectively. All had acute onset, presenting with progressive and severe muscle weakness. All lost ambulation within 1 or 2 months, with difficult swallowing and low voice. Respiratory distress occurred in case 2 after an attack of asphyxia due to an aspiration of sputum, and ventilator support was required for 1 month. Rashes were detected at the initial stage of the disease in cases 2 and 3. Patient 2 showed facial erythematous papules, spreading to her neck and hands. Patient 3 showed purplish eyelids with peri-orbital swelling, generalized edema involving all her limbs. Creatine kinase (CK) levels were markedly elevated in all the patients, ranging from 6 000 IU/L to 28 819 IU/L. Anti-SRP antibody was identified in cases 1, and anti-NXP2 antibodies were confirmed in cases 2 and 3. MRI of both thighs in all the patients showed profound muscle and fascial edema. Muscle pathology of patient 1 showed prominent fiber variation and endomysial fibrosis, with overexpression of MHC-Ⅰ. While muscle pathology in patients 2 and 3 showed scattered fiber necrosis, regeneration, endomysial edema without inflammatory cell infiltration. All the patients were diagnosed with idiopathic inflammatory myopathy and failed to the initial treatment including adequate glucocorticoids and high-dose immunoglobulin therapy. Other immunosuppressants (methotrexate, cyclophosphamide) were also tried in cases 2 and 3 with poor response. Then all the patients were treated with rituximab combined with glucocorticoids. Patient 1 regained normal strength and discontinued rituximab at the end of her last follow-up (2 years and 7 mouths). Though calcinosis developed during the follow-up period, significant improvement was noticed in cases 2 and 3 (both regained the ability to walk independently) at the end of their last follow-up after 2 years and 8 months, 3 years and 2 months respectively. Long-term rituximab therapy may improve the prognosis of refractory idiopathic inflammatory myopathy, especially with positive anti-SRP and anti-NXP2 antibodies.
Collapse
Affiliation(s)
- 文霞 伊
- 北京大学第一医院儿科,北京 100034Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
- 沧州市中心医院儿科,河北沧州 061000Department of Pediatrics, Cangzhou Central Hospital, Cangzhou 061000, Hebei, China
| | - 翠洁 魏
- 北京大学第一医院儿科,北京 100034Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| | - 晔 吴
- 北京大学第一医院儿科,北京 100034Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| | - 新华 包
- 北京大学第一医院儿科,北京 100034Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| | - 晖 熊
- 北京大学第一医院儿科,北京 100034Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| | - 杏芝 常
- 北京大学第一医院儿科,北京 100034Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| |
Collapse
|
35
|
伊 文, 魏 翠, 吴 晔, 包 新, 熊 晖, 常 杏. [Long-term rituximab treatment of refractory idiopathic inflammatory myopathy: A report of 3 cases]. BEIJING DA XUE XUE BAO. YI XUE BAN = JOURNAL OF PEKING UNIVERSITY. HEALTH SCIENCES 2021; 53:1191-1195. [PMID: 34916704 PMCID: PMC8695162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Indexed: 08/20/2024]
Abstract
Idiopathic inflammatory myopathies are a group of rare but serious diseases. The treatment of refractory idiopathic inflammatory myopathy is always challenging, especially in children. Three cases of refractory idiopathic inflammatory myopathy treated by rituximab were reported and discussed with the review of relevant literature. All were female with on-set age of 8 years and 6 months, 11 years and 7 months, 4 years and 2 months old, respectively. All had acute onset, presenting with progressive and severe muscle weakness. All lost ambulation within 1 or 2 months, with difficult swallowing and low voice. Respiratory distress occurred in case 2 after an attack of asphyxia due to an aspiration of sputum, and ventilator support was required for 1 month. Rashes were detected at the initial stage of the disease in cases 2 and 3. Patient 2 showed facial erythematous papules, spreading to her neck and hands. Patient 3 showed purplish eyelids with peri-orbital swelling, generalized edema involving all her limbs. Creatine kinase (CK) levels were markedly elevated in all the patients, ranging from 6 000 IU/L to 28 819 IU/L. Anti-SRP antibody was identified in cases 1, and anti-NXP2 antibodies were confirmed in cases 2 and 3. MRI of both thighs in all the patients showed profound muscle and fascial edema. Muscle pathology of patient 1 showed prominent fiber variation and endomysial fibrosis, with overexpression of MHC-Ⅰ. While muscle pathology in patients 2 and 3 showed scattered fiber necrosis, regeneration, endomysial edema without inflammatory cell infiltration. All the patients were diagnosed with idiopathic inflammatory myopathy and failed to the initial treatment including adequate glucocorticoids and high-dose immunoglobulin therapy. Other immunosuppressants (methotrexate, cyclophosphamide) were also tried in cases 2 and 3 with poor response. Then all the patients were treated with rituximab combined with glucocorticoids. Patient 1 regained normal strength and discontinued rituximab at the end of her last follow-up (2 years and 7 mouths). Though calcinosis developed during the follow-up period, significant improvement was noticed in cases 2 and 3 (both regained the ability to walk independently) at the end of their last follow-up after 2 years and 8 months, 3 years and 2 months respectively. Long-term rituximab therapy may improve the prognosis of refractory idiopathic inflammatory myopathy, especially with positive anti-SRP and anti-NXP2 antibodies.
Collapse
Affiliation(s)
- 文霞 伊
- 北京大学第一医院儿科,北京 100034Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
- 沧州市中心医院儿科,河北沧州 061000Department of Pediatrics, Cangzhou Central Hospital, Cangzhou 061000, Hebei, China
| | - 翠洁 魏
- 北京大学第一医院儿科,北京 100034Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| | - 晔 吴
- 北京大学第一医院儿科,北京 100034Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| | - 新华 包
- 北京大学第一医院儿科,北京 100034Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| | - 晖 熊
- 北京大学第一医院儿科,北京 100034Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| | - 杏芝 常
- 北京大学第一医院儿科,北京 100034Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| |
Collapse
|
36
|
Kobayashi T, Nakano T, Ogata H, Sato N, Yamaide F, Yamashita Y, Chikaraishi K, Hino M, Nishino I, Ichimura Y, Okiyama N, Hamada H. A 10-year-old girl with low-grade B cell lymphoma complicated by anti-nuclear matrix protein 2 autoantibody-positive juvenile dermatomyositis. Rheumatology (Oxford) 2021; 61:e143-e145. [PMID: 34897378 DOI: 10.1093/rheumatology/keab922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/25/2021] [Accepted: 12/03/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Toshiyuki Kobayashi
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Chiba, Japan
| | - Taiji Nakano
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Chiba, Japan
| | - Hitoshi Ogata
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Chiba, Japan
| | - Noriko Sato
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Chiba, Japan
| | - Fumiya Yamaide
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Chiba, Japan
| | - Yoshiharu Yamashita
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Chiba, Japan
| | - Koji Chikaraishi
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Chiba, Japan
| | - Moeko Hino
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Chiba, Japan
| | - Ichizo Nishino
- Department of Neuromuscular Research, National Institute of Neuroscience,National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Yuki Ichimura
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Naoko Okiyama
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hiromichi Hamada
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Chiba, Japan
| |
Collapse
|
37
|
Physical activity in idiopathic inflammatory myopathies: two intervention proposals based on literature review. Clin Rheumatol 2021; 41:593-615. [PMID: 34665346 DOI: 10.1007/s10067-021-05954-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/15/2021] [Accepted: 10/03/2021] [Indexed: 12/18/2022]
Abstract
Idiopathic inflammatory myopathies (IIM) are rare diseases affecting skeletal muscles and leading to progressive muscle weakness and disability. Thanks to the better understanding of their pathogenesis, the management of IIM has been noteworthily implemented in recent years. Current therapeutic strategies include pharmacological and non-pharmacological interventions, among which physical exercise represents a useful option, able to ameliorate disease activity without worsening muscle inflammation. The aim of this narrative review is therefore to provide an updated overview of the benefits of physical exercise in patients with IIM and to suggest plausible training programs to be applied in patients with dermatomyositis, polymyositis, necrotizing myopathy, and inclusion body myositis. In this regard, a combined strategy mixing aerobic and resistance exercises could positively affect the pro-inflammatory and metabolic pathways occurring in skeletal muscles, while promoting muscle fiber regeneration and repair.
Collapse
|
38
|
Ll Wilkinson MG, Deakin CT, Papadopoulou C, Eleftheriou D, Wedderburn LR. JAK inhibitors: a potential treatment for JDM in the context of the role of interferon-driven pathology. Pediatr Rheumatol Online J 2021; 19:146. [PMID: 34563217 PMCID: PMC8466894 DOI: 10.1186/s12969-021-00637-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/28/2021] [Indexed: 12/29/2022] Open
Abstract
Juvenile Idiopathic Inflammatory Myopathies (IIM) are a group of rare diseases that are heterogeneous in terms of pathology that can include proximal muscle weakness, associated skin changes and systemic involvement. Despite options for treatment, many patients continue to suffer resistant disease and lasting side-effects. Advances in the understanding of the immunopathology and genetics underlying IIM may specify new therapeutic targets, particularly where conventional treatment has not achieved a clinical response. An upregulated type I interferon signature is strongly associated with disease and could be a prime target for developing more specific therapeutics. There are multiple components of the IFN pathway that could be targeted for blockade therapy.Downstream of the cytokine receptor complexes are the Janus kinase-signal transducers and activators of transcription (JAK-STAT) pathway, which consists of JAK1-3, TYK2, and STAT1-6. Therapeutic inhibitors have been developed to target components of this pathway. Promising results have been observed in case studies reporting the use of the JAK inhibitors, Baricitinib, Tofacitinib and Ruxolitinib in the treatment of refractory Juvenile Dermatomyositis (JDM). There is still the question of safety and efficacy for the use of JAK inhibitors in JDM that need to be addressed by clinical trials. Here we review the future for the use of JAK inhibitors as a treatment for JDM.
Collapse
Affiliation(s)
- Meredyth G Ll Wilkinson
- Infection, Immunity and Inflammation Programme Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK.
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, University College London, London, UK.
- NIHR Biomedical Research Centre at GOSH, London, UK.
| | - Claire T Deakin
- Infection, Immunity and Inflammation Programme Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, University College London, London, UK
- NIHR Biomedical Research Centre at GOSH, London, UK
| | - Charalampia Papadopoulou
- Infection, Immunity and Inflammation Programme Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
- Rheumatology, Great Ormond Street Hospital, Great Ormond Street, London, UK
| | - Despina Eleftheriou
- Infection, Immunity and Inflammation Programme Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
- Rheumatology, Great Ormond Street Hospital, Great Ormond Street, London, UK
| | - Lucy R Wedderburn
- Infection, Immunity and Inflammation Programme Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, University College London, London, UK
- NIHR Biomedical Research Centre at GOSH, London, UK
- Rheumatology, Great Ormond Street Hospital, Great Ormond Street, London, UK
| |
Collapse
|
39
|
Nitiyarom R, Charuvanij S, Likasitwattanakul S, Thanoophunchai C, Wisuthsarewong W. Juvenile dermatomyositis in Thai children: Retrospective review of 30 cases from a tertiary care center. Indian J Dermatol Venereol Leprol 2021; 88:162-170. [PMID: 34491668 DOI: 10.25259/ijdvl_297_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 03/01/2021] [Indexed: 11/04/2022]
Abstract
BACKGROUND Juvenile dermatomyositis is a rare condition, but it is the most common idiopathic inflammatory myopathy in pediatric patients. AIM To study the clinical manifestations, investigations, treatment, clinical course, and outcomes of juvenile dermatomyositis in Thai children. METHOD This retrospective study included juvenile dermatomyositis patients treated at Siriraj Hospital, a 2,300-bed national tertiary referral center in Bangkok, Thailand, from 1994 to 2019. RESULTS Thirty patients (22 females and 8 males) were included with a female to male ratio of 2.7:1. Median age at diagnosis was 5.1 years (range, 2.6-14.8 years). Median duration of illness before diagnosis was 6.5 months (range, 0.3-84.0 months). Acute and subacute onset occurred in the majority of patients. Presenting symptoms included muscle weakness in 27/30 (90%), skin rash in 26/30 (86.7%), muscle pain in 17/26 (65.4%), and arthralgia in 4/18 (22.2%) of patients. Dermatologic examination revealed Gottron's rash, heliotrope rash, and periungual telangiectasia in 25/30 (83.3%), 21/30 (70.0%), and 15/24 (62.5%) of patients, respectively. Interestingly, scalp dermatitis was found in 8/21 (38.1%) of patients. The most commonly used treatment regimen in this series was a combination of prednisolone and methotrexate. During the median follow-up of 3.1 years (range, 0.0-18.5 years), only one-third of patients were seen to have monocyclic disease. Extraskeletal osteosarcoma at a previous lesion of calcinosis cutis was observed in one patient at 12 years after juvenile dermatomyositis onset. LIMITATIONS This was a retrospective single-center study, and our results may not be generalizable to other healthcare settings. Prospective multicenter studies are needed to confirm the findings of this study. CONCLUSION juvenile dermatomyositis usually poses a diagnostic and therapeutic challenge, which can be compounded by the ethnic variations in the clinical presentation, as observed in this study. Asian patients tend to present with acute or subacute onset of disease, and arthralgia and/or arthritis are less common than in Caucasian patients. Scalp dermatitis is not uncommon in pediatric juvenile dermatomyositis patients. An association between juvenile dermatomyositis and malignancy, though rare, can occur.
Collapse
Affiliation(s)
- Rattanavalai Nitiyarom
- Division of Dermatology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sirirat Charuvanij
- Division of Rheumatology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Surachai Likasitwattanakul
- Division of Neurology, Department of Pediatrics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chaiwat Thanoophunchai
- Division of Dermatology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Wanee Wisuthsarewong
- Division of Dermatology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
40
|
Lerkvaleekul B, Veldkamp SR, van der Wal MM, Schatorjé EJH, Kamphuis SSM, van den Berg JM, Muller PCEH, Armbrust W, Vastert SJ, Wienke J, Jansen MHA, van Royen-Kerkhof A, van Wijk F. Siglec-1 expression on monocytes is associated with the interferon signature in juvenile dermatomyositis and can predict treatment response. Rheumatology (Oxford) 2021; 61:2144-2155. [PMID: 34387304 PMCID: PMC9071568 DOI: 10.1093/rheumatology/keab601] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 07/20/2021] [Indexed: 12/01/2022] Open
Abstract
Objective JDM is a rare chronic immune-mediated inflammatory disease with a predominant role for type I IFN responses. We aimed to determine the potential of Siglec-1 expression on monocytes as a novel IFN-inducible biomarker for disease activity monitoring and prediction of treatment response in patients with JDM. Methods Siglec-1 was measured by flow cytometry on circulating monocytes of 21 newly diagnosed JDM patients before start of treatment and, for 10 of these, also during follow-up. The expression levels of five type I IFN-stimulated genes, MX1, IFI44, IFI44L, LY6E and IFIT3, were measured by RT-qPCR to determine the IFN signature and calculate an IFN score. IFN-inducible plasma proteins CXCL10 and galectin-9 were measured by multiplex immunoassay. Results Siglec-1 and IFN score were increased in JDM patients compared with controls and correlated with clinical disease activity. Stratification of patients by Siglec-1 expression at diagnosis identified those with high Siglec-1 expression as having a higher risk of requiring treatment intensification within the first 3 months after diagnosis (55% vs 0% of patients, P = 0.01). Siglec-1 expression strongly correlated with plasma levels of previously validated biomarkers CXCL10 (rs = 0.81, P < 0.0001) and galectin-9 (rs = 0.83, P < 0.0001), and was superior to the IFN score in predicting treatment response (area under the curve 0.87 vs 0.53, P = 0.01). Conclusion Siglec-1 on monocytes is a novel IFN-inducible biomarker in JDM that correlates with clinical disease activity and identifies patients at risk for a suboptimal treatment response. Further studies are required to validate these findings and their clinical potential.
Collapse
Affiliation(s)
- Butsabong Lerkvaleekul
- Division of Rheumatology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.,Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Saskia R Veldkamp
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Maria M van der Wal
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ellen J H Schatorjé
- Department of Paediatrics, Paediatric Rheumatology, Amalia Children's Hospital, Radboud University Medical Centre Nijmegen, Nijmegen, the Netherlands
| | - Sylvia S M Kamphuis
- Paediatric Rheumatology, Sophia Children's Hospital, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - J Merlijn van den Berg
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Petra C E Hissink Muller
- Department of Paediatric Rheumatology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Wineke Armbrust
- Department of Pediatric Rheumatology and Immunology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Sebastiaan J Vastert
- Pediatric Rheumatology and Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Judith Wienke
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marc H A Jansen
- Pediatric Rheumatology and Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Annet van Royen-Kerkhof
- Pediatric Rheumatology and Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Femke van Wijk
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
41
|
Deakin CT, Papadopoulou C, McCann LJ, Martin N, Al-Obaidi M, Compeyrot-Lacassagne S, Pilkington CA, Tansley SL, McHugh NJ, Wedderburn LR, De Stavola BL. Identification and prediction of novel classes of long-term disease trajectories for patients with juvenile dermatomyositis using growth mixture models. Rheumatology (Oxford) 2021; 60:1891-1901. [PMID: 33146389 PMCID: PMC8023987 DOI: 10.1093/rheumatology/keaa497] [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: 02/17/2020] [Revised: 07/11/2020] [Indexed: 11/13/2022] Open
Abstract
Objectives Uncertainty around clinical heterogeneity and outcomes for patients with JDM represents a major burden of disease and a challenge for clinical management. We sought to identify novel classes of patients having similar temporal patterns in disease activity and relate them to baseline clinical features. Methods Data were obtained for n = 519 patients, including baseline demographic and clinical features, baseline and follow-up records of physician’s global assessment of disease (PGA), and skin disease activity (modified DAS). Growth mixture models (GMMs) were fitted to identify classes of patients with similar trajectories of these variables. Baseline predictors of class membership were identified using Lasso regression. Results GMM analysis of PGA identified two classes of patients. Patients in class 1 (89%) tended to improve, while patients in class 2 (11%) had more persistent disease. Lasso regression identified abnormal respiration, lipodystrophy and time since diagnosis as baseline predictors of class 2 membership, with estimated odds ratios, controlling for the other two variables, of 1.91 for presence of abnormal respiration, 1.92 for lipodystrophy and 1.32 for time since diagnosis. GMM analysis of modified DAS identified three classes of patients. Patients in classes 1 (16%) and 2 (12%) had higher levels of modified DAS at diagnosis that improved or remained high, respectively. Patients in class 3 (72%) began with lower DAS levels that improved more quickly. Higher proportions of patients in PGA class 2 were in DAS class 2 (19%, compared with 16 and 10%). Conclusion GMM analysis identified novel JDM phenotypes based on longitudinal PGA and modified DAS.
Collapse
Affiliation(s)
- Claire T Deakin
- Infection, Immunity and Inflammation Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, UK.,Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCL Hospitals and Great Ormond Street Hospital, London, UK.,NIHR Biomedical Research Centre at Great Ormond Street Hospital, London, UK
| | - Charalampia Papadopoulou
- Infection, Immunity and Inflammation Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, UK.,Rheumatology Unit, Great Ormond Street Hospital, London, UK
| | - Liza J McCann
- Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust, Liverpool, UK
| | - Neil Martin
- Rheumatology Department, Royal Hospital for Sick Children, Glasgow
| | | | | | | | - Sarah L Tansley
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | - Neil J McHugh
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | - Lucy R Wedderburn
- Infection, Immunity and Inflammation Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, UK.,Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCL Hospitals and Great Ormond Street Hospital, London, UK.,NIHR Biomedical Research Centre at Great Ormond Street Hospital, London, UK.,Rheumatology Unit, Great Ormond Street Hospital, London, UK
| | - Bianca L De Stavola
- Population, Policy and Practice Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | | |
Collapse
|
42
|
Pauling JD, Christopher-Stine L. The aetiopathogenic significance, clinical relevance and therapeutic implications of vasculopathy in idiopathic inflammatory myopathy. Rheumatology (Oxford) 2021; 60:1593-1607. [PMID: 33458769 DOI: 10.1093/rheumatology/keaa816] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/16/2020] [Accepted: 10/15/2020] [Indexed: 12/31/2022] Open
Abstract
It is 120 years since 'angiomyositis' was included alongside 'polymyositis' and 'dermatomyositis' in an attempt to propose a taxonomy that reflected the major clinical characteristics of idiopathic inflammatory myopathy (IIM). Endothelial injury, perivascular inflammation and capillary loss are important histological findings in affected tissues in IIM. Overt vascular clinical features including RP and abnormal nailfold capillaroscopy (NC) are also common in IIM. Despite the presence of endothelial injury, perivascular inflammation and capillary loss in affected tissues in IIM, and the presence of clinical features such as RP and NC abnormalities, the pathogenic and therapeutic implications of vasculopathy in IIM have been somewhat overlooked. RP and NC abnormalities are not always present, providing a valuable opportunity to explore aetiopathogenic factors driving vasculopathy within autoimmune rheumatic disease. The present review examines the aetiopathogenic, prognostic and therapeutic significance of vasculopathy in IIM. We describe the prevalence and clinical relevance of vasculopathy in IIM, and consider how vasculopathy may be better utilized to support improved IIM diagnosis and disease classification. Areas of unmet research need are highlighted where relevant.
Collapse
Affiliation(s)
- John D Pauling
- Royal National Hospital for Rheumatic Diseases (Part of the Royal United Hospitals NHS Foundation Trust), Bath, UK.,Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | | |
Collapse
|
43
|
Mamyrova G, Kishi T, Shi M, Targoff IN, Huber AM, Curiel RV, Miller FW, Rider LG. Anti-MDA5 autoantibodies associated with juvenile dermatomyositis constitute a distinct phenotype in North America. Rheumatology (Oxford) 2021; 60:1839-1849. [PMID: 33140079 DOI: 10.1093/rheumatology/keaa429] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/03/2020] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE Myositis-specific autoantibodies have defined distinct phenotypes of patients with juvenile myositis (JIIM). We assessed the frequency and clinical significance of anti-melanoma differentiation-associated gene 5 (MDA5) autoantibody-associated JIIM in a North American registry. METHODS Retrospective examination of the characteristics of 35 JIIM patients with anti-MDA5 autoantibodies was performed, and differences from other myositis-specific autoantibody groups were evaluated. RESULTS Anti-MDA5 autoantibodies were present in 35/453 (7.7%) of JIIM patients and associated with older age at diagnosis, and lower serum creatine kinase and aldolase levels. Patients with anti-MDA5 autoantibodies had more frequent weight loss, adenopathy, arthritis, interstitial lung disease (ILD), and less frequent falling compared with anti-transcriptional intermediary factor 1 (TIF1), anti-nuclear matrix protein 2 (NXP2) and myositis-specific autoantibody/myositis-associated autoantibody-negative patients. They had a different season of diagnosis and less frequent mechanic's hands and ILD compared with those with anti-synthetase autoantibodies. Anti-MDA5 patients received fewer medications compared with anti-TIF1, and corticosteroid treatment was shorter compared with anti-TIF1 and anti-nuclear matrix protein 2 autoantibody groups. The frequency of remission was higher in anti-MDA5 than anti-synthetase autoantibody-positive JIIM. In multivariable analyses, weight loss, arthritis and arthralgia were most strongly associated with anti-MDA5 autoantibody-positive JIIM. CONCLUSION Anti-MDA5 JIIM is a distinct subset, with frequent arthritis, weight loss, adenopathy and less severe myositis, and is also associated with ILD. Anti-MDA5 is distinguished from anti-synthetase autoantibody-positive JIIM by less frequent ILD, lower creatine kinase levels and differing seasons of diagnosis. Anti-MDA5 has comparable outcomes, but with the ability to discontinue steroids more rapidly and less frequent flares compared with anti-TIF1 autoantibodies, and more frequent remission compared with anti-synthetase JIIM patients.
Collapse
Affiliation(s)
- Gulnara Mamyrova
- Division of Rheumatology, Department of Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Takayuki Kishi
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Min Shi
- Biostatistics & Computational Biology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Ira N Targoff
- Veteran's Affairs Medical Center, University of Oklahoma Health Sciences Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Adam M Huber
- IWK Health Centre and Dalhousie University, Halifax, NS, Canada
| | - Rodolfo V Curiel
- Division of Rheumatology, Department of Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Frederick W Miller
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Lisa G Rider
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, MD, USA
| | | |
Collapse
|
44
|
Kishi T, Warren-Hicks W, Bayat N, Targoff IN, Huber AM, Ward MM, Rider LG. Corticosteroid discontinuation, complete clinical response and remission in juvenile dermatomyositis. Rheumatology (Oxford) 2021; 60:2134-2145. [PMID: 33067611 DOI: 10.1093/rheumatology/keaa371] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/31/2020] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE A North American registry of JDM patients was examined for frequency of and factors associated with corticosteroid discontinuation, complete clinical response and remission. METHODS We evaluated probability of achieving final corticosteroid discontinuation, complete clinical response and remission in 307 JDM patients by Weibull time-to-event modelling; conditional probability of complete clinical response and remission using Bayesian network modelling; and significant predictors with multivariable Markov chain Monte-Carlo Weibull extension models. RESULTS The probability of corticosteroid discontinuation was 56%, complete clinical response 38% and remission 30% by 60 months after initial treatment in 105 patients. The probability of remission was conditional on corticosteroid discontinuation and complete clinical response. Photosensitivity, contractures and a longer time to complete clinical response were predictive of the time to final corticosteroid discontinuation. Anti-MJ (NXP2) autoantibodies and a Northwest residential geoclimatic zone were predictive of shorter time to complete clinical response, while dysphonia, contractures, an increase in medications within 24 months and a longer time to corticosteroid discontinuation were associated with longer time to complete clinical response. Anti-p155/140 (TIF1) autoantibodies, an increase in medications within 12-24 months, or longer times to corticosteroid discontinuation and complete clinical response were associated with longer time to remission. CONCLUSION JDM patients achieve favourable outcomes, including corticosteroid discontinuation, complete clinical response and remission, although timelines for these may be several years based on time-dependent analyses. These outcomes are inter-related and strong predictors of each other. Selected clinical features and myositis autoantibodies are additionally associated with these outcomes.
Collapse
Affiliation(s)
- Takayuki Kishi
- Environmental Autoimmunity Group, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, MD
| | | | - Nastaran Bayat
- Environmental Autoimmunity Group, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, MD
| | - Ira N Targoff
- VA Medical Center, University of Oklahoma Health Sciences Center.,Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Adam M Huber
- IWK Health Centre and Dalhousie University, Halifax, Nova Scotia, Canada
| | - Michael M Ward
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lisa G Rider
- Environmental Autoimmunity Group, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, MD
| | | |
Collapse
|
45
|
Neely J, Shalen J, Sturrock H, Kim S. Access to Care and Diagnostic Delays in Juvenile Dermatomyositis: Results From the Childhood Arthritis and Rheumatology Research Alliance Legacy Registry. ACR Open Rheumatol 2021; 3:349-354. [PMID: 33932146 PMCID: PMC8126761 DOI: 10.1002/acr2.11246] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 02/10/2021] [Indexed: 11/09/2022] Open
Abstract
Objective To determine factors associated with diagnostic delays and outcomes in juvenile dermatomyositis (JDM) in the Childhood Arthritis and Rheumatology Research Alliance Legacy Registry (CLR). Methods This was a cross‐sectional study of subjects aged 0 to 17 years with JDM enrolled to the CLR from 2010 to 2015. Access to care was measured by calculating the distance from the subject zip code of residence to the treating pediatric rheumatology center and determining the state density of pediatric rheumatologists based on the 2015 American College of Rheumatology Workforce Study. Delay was categorized as early (<30 days), typical (1‐3 months), moderate (3‐12 months), and severe (>12 months). Ordered generalized additive models were used to determine the association between these measures and diagnostic delays. Results The median time to diagnosis was 3.1 months; 37.2% of patients experienced moderate delays, and 14.6% experienced severe delays. In a univariate analysis, younger age of disease onset and male sex were associated with delays. Using a generalized additive model accounting for age, sex, race, and ethnicity, increasing distance from treating pediatric rheumatologist and younger age at disease onset were associated with diagnostic delay. There was no association between the state density of rheumatologists and diagnostic delays in this model. Conclusion In the CLR, we found moderate to severe diagnostic delays in the majority of subjects with JDM. Our data suggest that access to care, measured as the distance traveled to treating rheumatologist, is an important factor associated with delays in care but also highlight age as a contributing factor, suggesting that JDM may be less recognizable in young children.
Collapse
Affiliation(s)
| | | | | | - Susan Kim
- University of California, San Francisco
| | | |
Collapse
|
46
|
Aguilar-Vazquez A, Chavarria-Avila E, Pizano-Martinez O, Ramos-Hernandez A, Andrade-Ortega L, Rubio-Arellano ED, Vazquez-Del Mercado M. Geographical Latitude Remains as an Important Factor for the Prevalence of Some Myositis Autoantibodies: A Systematic Review. Front Immunol 2021; 12:672008. [PMID: 33968081 PMCID: PMC8100663 DOI: 10.3389/fimmu.2021.672008] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/06/2021] [Indexed: 11/13/2022] Open
Abstract
The idiopathic inflammatory myopathies (IIM) are characterized by muscular weakness, cutaneous manifestations, muscle damage revealed by increase of muscular enzymes, muscle biopsy, electromyography and changes on magnetic resonance imaging. However, the hallmark of these IIM, is the development of myositis specific antibodies (MSA) or myositis associated antibodies (MAA). The theories about their presence in the serum of IIM is not known. Some studies have suggested that some of these MSA, such as anti-Mi-2 increases according to the intensity of UV radiation. There is scarce information about the environmental factors that might contribute in order to be considered as triggering factors as UV radiation might be. In this review, we analyzed the reported prevalence of MSAs and MAAs regarding to their geographical location and the possible relation with UV radiation. We collected the prevalence data of fifteen MSA and thirteen MAA from 22 countries around the world and we were able to observe a difference in prevalence between countries and continents. We found differences in anti-PL7, anti-Ro52, anti-La and anti-Ku prevalence according to UV radiation level. Otherwise, we observed that anti-Mi-2 prevalence increases near to the Equator meanwhile anti-MJ/NXP2 and anti-ARS prevalence had an opposite behavior increasing their prevalence in the geographical locations farther to the Equator. Our results highlighted the importance to include the UV radiation and other environmental factors in IIM studies, in order to clarify its association with MSA and MAA prevalence as well as its possible role in the immunopathogenesis of these diseases.
Collapse
Affiliation(s)
- Andrea Aguilar-Vazquez
- Centro Universitario de Ciencias de la Salud, Doctorado en Ciencias Biomédicas, Universidad de Guadalajara, Guadalajara, Mexico.,Centro Universitario de Ciencias de la Salud, Instituto de Investigación en Reumatología y del Sistema Músculo-Esquelético (IIRSME), Universidad de Guadalajara, Guadalajara, Mexico
| | - Efrain Chavarria-Avila
- Centro Universitario de Ciencias de la Salud, Instituto de Investigación en Reumatología y del Sistema Músculo-Esquelético (IIRSME), Universidad de Guadalajara, Guadalajara, Mexico.,Centro Universitario de Ciencias de la Salud, Departamento de Disciplinas Filosófico, Metodológicas e Instrumentales, Universidad de Guadalajara, Guadalajara, Mexico.,Hospital Civil Dr. Juan I. Menchaca, División de Medicina Interna, Servicio de Reumatología 004086, PNPC CONACyT, Guadalajara, Mexico
| | - Oscar Pizano-Martinez
- Centro Universitario de Ciencias de la Salud, Instituto de Investigación en Reumatología y del Sistema Músculo-Esquelético (IIRSME), Universidad de Guadalajara, Guadalajara, Mexico.,Hospital Civil Dr. Juan I. Menchaca, División de Medicina Interna, Servicio de Reumatología 004086, PNPC CONACyT, Guadalajara, Mexico.,Centro Universitario de Ciencias de la Salud, UDG-CA 703 Inmunología y Reumatología, Universidad de Guadalajara, Guadalajara, Mexico
| | - Alejandra Ramos-Hernandez
- Hospital Civil Dr. Juan I. Menchaca, División de Medicina Interna, Servicio de Reumatología 004086, PNPC CONACyT, Guadalajara, Mexico
| | - Lilia Andrade-Ortega
- Departamento de Reumatología Centro Médico Nacional 20 de Noviembre, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado (ISSSTE), Ciudad de México, Mexico
| | - Edy-David Rubio-Arellano
- Centro Universitario de Ciencias de la Salud, Departamento de Fisiología, Universidad de Guadalajara, Guadalajara, Mexico
| | - Monica Vazquez-Del Mercado
- Centro Universitario de Ciencias de la Salud, Instituto de Investigación en Reumatología y del Sistema Músculo-Esquelético (IIRSME), Universidad de Guadalajara, Guadalajara, Mexico.,Hospital Civil Dr. Juan I. Menchaca, División de Medicina Interna, Servicio de Reumatología 004086, PNPC CONACyT, Guadalajara, Mexico.,Centro Universitario de Ciencias de la Salud, UDG-CA 703 Inmunología y Reumatología, Universidad de Guadalajara, Guadalajara, Mexico.,Centro Universitario de Ciencias de la Salud, Departamento de Biología Molecular y Genómica, Universidad de Guadalajara, Guadalajara, Mexico
| |
Collapse
|
47
|
Sabbagh SE, Pinal-Fernandez I, Casal-Dominguez M, Albayda J, Paik JJ, Miller FW, Rider LG, Mammen AL, Christopher-Stine L. Anti-mitochondrial autoantibodies are associated with cardiomyopathy, dysphagia, and features of more severe disease in adult-onset myositis. Clin Rheumatol 2021; 40:4095-4100. [PMID: 33851273 PMCID: PMC8463345 DOI: 10.1007/s10067-021-05730-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 11/25/2022]
Abstract
We analyzed the prevalence of anti-mitochondrial autoantibodies (AMA) in adult- and juvenile-onset myositis longitudinal cohorts and investigated phenotypic differences in myositis patients with AMA. We screened sera from myositis patients including 619 adult- and 371 juvenile-onset dermatomyositis (DM, JDM), polymyositis (PM, JPM), inclusion body myositis (IBM), or amyopathic DM patients and from healthy controls, including 164 adults and 92 children, for AMA by ELISA. Clinical characteristics were compared between myositis patients with and without AMA. AMA were present in 5% of adult myositis patients (16 of 216 DM, 10 of 222 PM, 4 of 140 IBM, 1 of 19 amyopathic DM), 1% of juvenile myositis patients (3 of 302 JDM, 1 of 25 JPM), and 1% of both adult and juvenile healthy controls. In patients with adult-onset myositis, AMA were associated with persistent muscle weakness, Raynaud’s phenomenon, dysphagia, and cardiomyopathy. Adult myositis patients with AMA may have more severe or treatment refractory disease, as they more frequently received glucocorticoids and intravenous immunoglobulin. In juvenile myositis, children with AMA often had falling episodes and dysphagia, but no other clinical features or medications were significantly associated with AMA. AMA are present in 5% of adult myositis patients and associated with cardiomyopathy, dysphagia, and other signs of severe disease. The prevalence of AMA is not increased in patients with juvenile myositis compared to age-matched healthy controls. Our data suggest that the presence of AMA in adult myositis patients should prompt screening for cardiac and swallowing involvement.
Key Points • Approximately 5% of a large North American cohort of adult myositis patients have anti-mitochondrial autoantibodies. • Adults with anti-mitochondrial autoantibodies often have chronic weakness, Raynaud’s, dysphagia, cardiomyopathy, and more severe disease. • Anti-mitochondrial autoantibodies are rare in juvenile myositis and not associated with a specific clinical phenotype. |
Collapse
Affiliation(s)
- Sara E Sabbagh
- Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA.,Division of Rheumatology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Iago Pinal-Fernandez
- Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Faculty of Health Sciences, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Maria Casal-Dominguez
- Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jemima Albayda
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Julie J Paik
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Frederick W Miller
- Environmental Autoimmunity Group, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Lisa G Rider
- Environmental Autoimmunity Group, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Andrew L Mammen
- Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lisa Christopher-Stine
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Johns Hopkins Myositis Center, Division of Rheumatology, Johns Hopkins University School of Medicine, Bayview Medical Office, 5200 Eastern Ave #301, Baltimore, MD, 21224, USA.
| | | |
Collapse
|
48
|
Bhattarai D, Jindal AK, Patra PK. Hypopigmentation in the Medial Angle of Eyes in a Young Boy With Juvenile Dermatomyositis. J Clin Rheumatol 2021; 27:e120. [PMID: 31977652 DOI: 10.1097/rhu.0000000000001294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Dharmagat Bhattarai
- From the Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | | | | |
Collapse
|
49
|
Anti-SAE autoantibody-positive Japanese patient with juvenile dermatomyositis complicated with interstitial lung disease - a case report. Pediatr Rheumatol Online J 2021; 19:34. [PMID: 33740993 PMCID: PMC7980636 DOI: 10.1186/s12969-021-00532-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 03/10/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Clinical phenotypes and outcomes in juvenile dermatomyositis (JDM) have been defined by various myositis-specific autoantibodies (MSAs). One of the recently described MSAs associated with DM is targeted against the small ubiquitin-like modifier 1 activating enzyme (SAE). We report an anti-SAE autoantibody-positive JDM patient complicated with interstitial lung disease (ILD). CASE PRESENTATION An 8-year-8-month-old Japanese girl presented with bilateral eyelid edema and facial erythema. At 8 years 4 months, she had dry cough and papules with erythema on the dorsal side of the interphalangeal joints of both hands. Her facial erythema gradually worsened and did not improve with topical steroids. At the first visit to our department at 8 years 8 months of age, she had a typical heliotrope rash and Gottron's papules, with no fever or weight loss, and a chest computed tomography scan showed ground-glass opacity under visceral pleura. There was no clinical evidence of myositis, muscle weakness, myalgia, or muscle magnetic resonance imaging (MRI) findings. She had mild dry cough, without any signs of respiratory distress. Laboratory tests showed no elevated inflammatory markers. She had a normal serum creatine kinase level with a slightly elevated aldolase level, and serum anti-SAE autoantibody was detected by immunoprecipitation-western blotting. She was diagnosed with juvenile amyopathic DM complicated by ILD and received two courses of methylprednisolone pulse therapy followed by oral corticosteroid and cyclosporin A. We gradually reduced the corticosteroid dose as her skin rash improved after treatment initiation. There was no progression of muscle symptoms, dysphagia, or disease flare during a 24-month follow-up period. CONCLUSIONS We report a patient with anti-SAE autoantibody-positive JDM complicated by interstitial pneumonia. This patient had no progression of muscle symptoms and dysphagia during a 24-month follow-up period, which differs from previous reports in adult patients with MSAs. There have been no previous reports of pediatric patients with SAE presenting with ILD. However, ILD seen in this case was not rapidly progressive and did not require cytotoxic agents. To prevent overtreatment, appropriate treatment choices are required considering the type of ILD.
Collapse
|
50
|
Fang C, Xu D, Su J, Dry JR, Linghu B. DeePaN: deep patient graph convolutional network integrating clinico-genomic evidence to stratify lung cancers for immunotherapy. NPJ Digit Med 2021; 4:14. [PMID: 33531613 PMCID: PMC7854753 DOI: 10.1038/s41746-021-00381-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 01/05/2021] [Indexed: 01/30/2023] Open
Abstract
Immuno-oncology (IO) therapies have transformed the therapeutic landscape of non-small cell lung cancer (NSCLC). However, patient responses to IO are variable and influenced by a heterogeneous combination of health, immune, and tumor factors. There is a pressing need to discover the distinct NSCLC subgroups that influence response. We have developed a deep patient graph convolutional network, we call "DeePaN", to discover NSCLC complexity across data modalities impacting IO benefit. DeePaN employs high-dimensional data derived from both real-world evidence (RWE)-based electronic health records (EHRs) and genomics across 1937 IO-treated NSCLC patients. DeePaN demonstrated effectiveness to stratify patients into subgroups with significantly different (P-value of 2.2 × 10-11) overall median survival of 20.35 months and 9.42 months post-IO therapy. Significant differences in IO outcome were not seen from multiple non-graph-based unsupervised methods. Furthermore, we demonstrate that patient stratification from DeePaN has the potential to augment the emerging IO biomarker of tumor mutation burden (TMB). Characterization of the subgroups discovered by DeePaN indicates potential to inform IO therapeutic insight, including the enrichment of mutated KRAS and high blood monocyte count in the IO beneficial and IO non-beneficial subgroups, respectively. Our work has proven the concept that graph-based AI is feasible and can effectively integrate high-dimensional genomic and EHR data to meaningfully stratify cancer patients on distinct clinical outcomes, with potential to inform precision oncology.
Collapse
Affiliation(s)
- Chao Fang
- Translational Medicine, Research and Early Development, Oncology R&D, AstraZeneca, Boston, MA, USA
| | - Dong Xu
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, USA
- Christopher S Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Jing Su
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN, USA.
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA.
| | - Jonathan R Dry
- Translational Medicine, Research and Early Development, Oncology R&D, AstraZeneca, Boston, MA, USA.
| | - Bolan Linghu
- Translational Medicine, Research and Early Development, Oncology R&D, AstraZeneca, Boston, MA, USA.
| |
Collapse
|