1
|
Martín-Torregrosa D, Mansilla-Polo M, Morgado-Carrasco D. Use of Anifrolumab in Systemic Lupus Erythematosus, Cutaneous Lupus Erythematosus, and Other Autoimmune Dermatoses. ACTAS DERMO-SIFILIOGRAFICAS 2025; 116:55-67. [PMID: 38972582 DOI: 10.1016/j.ad.2024.05.024] [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: 03/22/2024] [Revised: 05/13/2024] [Accepted: 05/21/2024] [Indexed: 07/09/2024] Open
Abstract
Anifrolumab is an inhibitor of the type I interferon receptor subunit 1 (IFNAR1) recently approved for the management of moderate-to-severe systemic lupus erythematosus (SLE). In 2 clinical trials, it has proven effective to treat cutaneous signs. Although anifrolumab has not been indicated for cutaneous lupus erythematosus (CLE), multiple cases and case series (20 publications with a total of 78 patients) have shown good and rapid responses with this drug, both in subacute CLE and discoid lupus erythematosus, as well as in lupus panniculitis and perniosis. Two case reports of dermatomyositis have also experienced clinical improvement with anifrolumab. Clinical trials of this drug are ongoing for subacute CLE and discoid lupus erythematosus, systemic sclerosis, and progressive vitiligo. Its most common adverse effects are respiratory infections and herpes zoster. Anifrolumab may be a well-tolerated alternative in the management of CLE.
Collapse
Affiliation(s)
- D Martín-Torregrosa
- Servicio de Dermatología, Hospital Universitario y Politécnico La Fe, IIS La Fe, Valencia, España
| | - M Mansilla-Polo
- Servicio de Dermatología, Hospital Universitario y Politécnico La Fe, IIS La Fe, Valencia, España
| | - D Morgado-Carrasco
- Servicio de Dermatología, Hospital de Figueres, Fundació Salut Empordà, Girona, España; Servicio de Dermatología, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, España.
| |
Collapse
|
2
|
Martín-Torregrosa D, Mansilla-Polo M, Morgado-Carrasco D. [Translated article] Use of Anifrolumab in Systemic Lupus Erythematosus, Cutaneous Lupus Erythematosus, and Other Autoimmune Dermatoses. ACTAS DERMO-SIFILIOGRAFICAS 2025; 116:T55-T67. [PMID: 39389344 DOI: 10.1016/j.ad.2024.10.008] [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: 03/22/2024] [Revised: 05/13/2024] [Accepted: 05/21/2024] [Indexed: 10/12/2024] Open
Abstract
Anifrolumab is an inhibitor of the type I interferon receptor subunit 1 (IFNAR1) recently approved for the management of moderate-to-severe systemic lupus erythematosus (SLE). In 2 clinical trials, it has proven effective to treat cutaneous signs. Although anifrolumab has not been indicated for cutaneous lupus erythematosus (CLE), multiple cases and case series (20 publications with a total of 78 patients) have shown good and rapid responses with this drug, both in subacute CLE and discoid lupus erythematosus, as well as in lupus panniculitis and perniosis. Two case reports of dermatomyositis have also experienced clinical improvement with anifrolumab. Clinical trials of this drug are ongoing for subacute CLE and discoid lupus erythematosus, systemic sclerosis, and progressive vitiligo. Its most common adverse effects are respiratory infections and herpes zoster. Anifrolumab may be a well-tolerated alternative in the management of CLE.
Collapse
MESH Headings
- Humans
- Lupus Erythematosus, Cutaneous/chemically induced
- Lupus Erythematosus, Cutaneous/drug therapy
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Lupus Erythematosus, Systemic/drug therapy
- Lupus Erythematosus, Systemic/chemically induced
- Lupus Erythematosus, Systemic/complications
- Autoimmune Diseases/drug therapy
- Autoimmune Diseases/chemically induced
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal/adverse effects
- Skin Diseases/chemically induced
- Clinical Trials as Topic
Collapse
Affiliation(s)
- D Martín-Torregrosa
- Servicio de Dermatología, Hospital Universitario y Politécnico La Fe, IIS La Fe, Valencia, Spain
| | - M Mansilla-Polo
- Servicio de Dermatología, Hospital Universitario y Politécnico La Fe, IIS La Fe, Valencia, Spain
| | - D Morgado-Carrasco
- Servicio de Dermatología, Hospital de Figueres, Fundació Salut Empordà, Girona, Spain; Servicio de Dermatología, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
3
|
Arkin LM, Costa-da-Silva AC, Frere J, Ng A, Sharma R, Moon JJ, Bussan HE, Kim CH, Javaid A, Steidl OR, Yatim A, Saidoune F, Gilliet M, Nguyen JT, Nihal A, Luong G, Kenfield M, Carrau L, Tran JM, Hinshaw MA, Brooks EG, Ayuso JM, O'Connor DH, Casanova JL, Cowen EW, Drolet BA, Singh AM, tenOever B, Mays JW. Pandemic-associated pernio harbors footprints of an abortive SARS-CoV-2 infection. iScience 2024; 27:110525. [PMID: 39156641 PMCID: PMC11326933 DOI: 10.1016/j.isci.2024.110525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 05/31/2024] [Accepted: 07/12/2024] [Indexed: 08/20/2024] Open
Abstract
Elevated pernio incidence was observed during the COVID-19 pandemic. This prospective study enrolled subjects with pandemic-associated pernio in Wisconsin and Switzerland. Because pernio is a cutaneous manifestation of the interferonopathies, and type I interferon (IFN-I) immunity is critical to COVID-19 recovery, we tested the hypothesis that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-mediated IFN-I signaling might underlie some pernio cases. Tissue-level IFN-I activity and plasmacytoid dendritic cell infiltrates were demonstrated in 100% of the Wisconsin cases. Across both cohorts, sparse SARS-CoV-2 RNA was captured in 25% (6/22) of biopsies, all with high inflammation. Affected patients lacked adaptive immunity to SARS-CoV-2. A hamster model of intranasal SARS-CoV-2 infection was used as a proof-of-principle experiment: RNA was detected in lungs and toes with IFN-I activity at both the sites, while replicating virus was found only in the lung. These data support a viral trigger for some pernio cases, where sustained local IFN-I activity can be triggered in the absence of seroconversion.
Collapse
Affiliation(s)
- Lisa M. Arkin
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Dermatology, Madison, WI 53726, USA
| | - Ana C. Costa-da-Silva
- Oral Immunobiology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Justin Frere
- Department of Microbiology, New York University, Grossman School of Medicine, New York, NY 10016, USA
| | - Ashley Ng
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Dermatology, Madison, WI 53726, USA
| | - Rubina Sharma
- Oral Immunobiology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - John J. Moon
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Dermatology, Madison, WI 53726, USA
| | - Hailey E. Bussan
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Pathology and Laboratory Medicine, Madison, WI 53726, USA
| | - Clara H. Kim
- Oral Immunobiology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ayesha Javaid
- Oral Immunobiology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Olivia R. Steidl
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Pediatrics, Madison, WI 53726, USA
| | - Ahmad Yatim
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
- Department of Dermatology at the University Hospital CHUV, 1011 Lausanne, Switzerland
| | - Fanny Saidoune
- Department of Dermatology at the University Hospital CHUV, 1011 Lausanne, Switzerland
| | - Michel Gilliet
- Department of Dermatology at the University Hospital CHUV, 1011 Lausanne, Switzerland
| | - Joe T. Nguyen
- Oral Immunobiology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Aman Nihal
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Dermatology, Madison, WI 53726, USA
| | - George Luong
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Dermatology, Madison, WI 53726, USA
| | - Meaghan Kenfield
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Dermatology, Madison, WI 53726, USA
| | - Lucia Carrau
- Department of Microbiology, New York University, Grossman School of Medicine, New York, NY 10016, USA
| | - Jennifer M. Tran
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Dermatology, Madison, WI 53726, USA
| | - Molly A. Hinshaw
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Dermatology, Madison, WI 53726, USA
| | - Erin G. Brooks
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Pathology and Laboratory Medicine, Madison, WI 53726, USA
| | - Jose M. Ayuso
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Dermatology, Madison, WI 53726, USA
| | - David H. O'Connor
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Pathology and Laboratory Medicine, Madison, WI 53726, USA
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
- University of Paris Cité, Imagine Institute, 75013 Paris, France
- Howard Hughes Medical Institute, New York, NY 10065, USA
- Lab of Human Genetics of Infectious Diseases, INSERM, Necker Hospital for Sick Children, Paris, France
- Department of Pediatrics, Necker Hospital for Sick Children, 75015 Paris, France
| | - Edward W. Cowen
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Beth A. Drolet
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Dermatology, Madison, WI 53726, USA
| | - Anne Marie Singh
- School of Medicine and Public Health, University of Wisconsin-Madison, Department of Pediatrics, Madison, WI 53726, USA
| | - Benjamin tenOever
- Department of Microbiology, New York University, Grossman School of Medicine, New York, NY 10016, USA
| | - Jacqueline W. Mays
- Oral Immunobiology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
4
|
Tanaka Y, Kusuda M, Yamaguchi Y. Interferons and systemic lupus erythematosus: Pathogenesis, clinical features, and treatments in interferon-driven disease. Mod Rheumatol 2023; 33:857-867. [PMID: 36440704 DOI: 10.1093/mr/roac140] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/24/2022] [Accepted: 11/09/2022] [Indexed: 08/27/2023]
Abstract
Type I interferons (IFNs) have recently received a lot of attention with the elucidation of the pathogenesis of systemic lupus erythematosus (SLE). Type I IFNs are associated with many SLE symptoms and play a role in the pathogenesis of autoimmune diseases that may occur concurrently with SLE, such as Sjögren's syndrome, antiphospholipid syndrome, myositis, scleroderma, and interferonopathy. Type I IFNs could be the link between these diseases. However, direct measurement of type I IFN levels and the IFN gene signature is currently unavailable in clinical practice. This review discusses type I IFN signalling in SLE, investigates the role of type I IFN in the clinical manifestations and symptoms associated with SLE and other IFN-related diseases, and discusses the clinical tests that can be used to diagnose SLE and measure disease activity. In addition, the role of type I IFN-blocking therapies as potential treatments for SLE is discussed.
Collapse
Affiliation(s)
- Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | | | | |
Collapse
|
5
|
Fetter T, de Graaf DM, Claus I, Wenzel J. Aberrant inflammasome activation as a driving force of human autoimmune skin disease. Front Immunol 2023; 14:1190388. [PMID: 37325658 PMCID: PMC10266227 DOI: 10.3389/fimmu.2023.1190388] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/09/2023] [Indexed: 06/17/2023] Open
Abstract
Autoimmune skin diseases are understood as conditions in which the adaptive immune system with autoantigen-specific T cells and autoantibody-producing B cells reacting against self-tissues plays a crucial pathogenic role. However, there is increasing evidence that inflammasomes, which are large multiprotein complexes that were first described 20 years ago, contribute to autoimmune disease progression. The inflammasome and its contribution to the bioactivation of interleukins IL-1β and IL-18 play an essential role in combating foreign pathogens or tissue damage, but may also act as a pathogenic driver of myriad chronic inflammatory diseases when dysfunctionally regulated. Inflammasomes containing the NOD-like receptor family members NLRP1 and NLRP3 as well as the AIM2-like receptor family member AIM2 have been increasingly investigated in inflammatory skin conditions. In addition to autoinflammatory diseases, which are often associated with skin involvement, the aberrant activation of the inflammasome has also been implied in autoimmune diseases that can either affect the skin besides other organs such as systemic lupus erythematosus and systemic sclerosis or are isolated to the skin in humans. The latter include, among others, the T-cell mediated disorders vitiligo, alopecia areata, lichen planus and cutaneous lupus erythematosus as well as the autoantibody-driven blistering skin disease bullous pemphigoid. Some diseases are characterized by both autoinflammatory and autoimmune responses such as the chronic inflammatory skin disease psoriasis. Further insights into inflammasome dysregulation and associated pathways as well as their role in forming adaptive immune responses in human autoimmune skin pathology could potentially offer a new field of therapeutic options in the future.
Collapse
Affiliation(s)
- Tanja Fetter
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | | | - Isabelle Claus
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Joerg Wenzel
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
6
|
Gleave A, Granville DJ. Granzyme B in Autoimmune Skin Disease. Biomolecules 2023; 13:388. [PMID: 36830757 PMCID: PMC9952967 DOI: 10.3390/biom13020388] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Autoimmune diseases often present with cutaneous symptoms that contribute to dysfunction, disfigurement, and in many cases, reduced quality-of-life. Unfortunately, treatment options for many autoimmune skin diseases are limited. Local and systemic corticosteroids remain the current standard-of-care but are associated with significant adverse effects. Hence, there is an unmet need for novel therapies that block molecular drivers of disease in a local and/or targeted manner. Granzyme B (GzmB) is a serine protease with known cytotoxic activity and emerging extracellular functions, including the cleavage of cell-cell junctions, basement membranes, cell receptors, and other structural proteins. While minimal to absent in healthy skin, GzmB is markedly elevated in alopecia areata, interface dermatitis, pemphigoid disease, psoriasis, systemic sclerosis, and vitiligo. This review will discuss the role of GzmB in immunity, blistering, apoptosis, and barrier dysfunction in the context of autoimmune skin disease. GzmB plays a causal role in the development of pemphigoid disease and carries diagnostic and prognostic significance in cutaneous lupus erythematosus, vitiligo, and alopecia areata. Taken together, these data support GzmB as a promising therapeutic target for autoimmune skin diseases impacted by impaired barrier function, inflammation, and/or blistering.
Collapse
Affiliation(s)
- Anna Gleave
- British Columbia Professional Firefighters’ Burn and Wound Healing Laboratory, International Collaboration On Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - David J. Granville
- British Columbia Professional Firefighters’ Burn and Wound Healing Laboratory, International Collaboration On Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada
| |
Collapse
|
7
|
Hernández Arroyo J, Izquierdo-Condoy JS, Ortiz-Prado E. A Case Series and Literature Review of Telogen Effluvium and Alopecia Universalis after the Administration of a Heterologous COVID-19 Vaccine Scheme. Vaccines (Basel) 2023; 11:vaccines11020444. [PMID: 36851320 PMCID: PMC9968192 DOI: 10.3390/vaccines11020444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 01/29/2023] [Accepted: 02/03/2023] [Indexed: 02/17/2023] Open
Abstract
COVID-19 vaccines have positively changed the course of the pandemic. They entered the market after only one year of the initial trials, which that yielded positive results in terms of safety and efficacy. However, after inoculating billions of people in the most extensive vaccination campaign worldwide, mild but common and some rare but potentially fatal adverse events have been reported. Among several self-reported adverse events, hair loss and alopecia have been linked to COVID-19 mRNA or viral vector vaccines. We tracked and followed a series of five cases with post-vaccine telogen effluvium and alopecia development in Ecuador. Here, we reported the clinical presentation of two women and three men with the diagnosis of post-vaccine hair loss. All patients received a heterologous vaccination scheme (mRNA and attenuated virus vaccine) with an additional viral vector booster associated with the apparition of telogen effluvium and alopecia universalis between 3 and 17 days after the vaccine was administered.
Collapse
Affiliation(s)
| | - Juan S. Izquierdo-Condoy
- One Health Research Group, Faculty of Medicine, Universidad de las Américas, Quito 170137, Ecuador
| | - Esteban Ortiz-Prado
- One Health Research Group, Faculty of Medicine, Universidad de las Américas, Quito 170137, Ecuador
- Correspondence: ; Tel.: +593-995760693
| |
Collapse
|
8
|
Sánchez-Pellicer P, Navarro-Moratalla L, Núñez-Delegido E, Agüera-Santos J, Navarro-López V. How Our Microbiome Influences the Pathogenesis of Alopecia Areata. Genes (Basel) 2022; 13:genes13101860. [PMID: 36292745 PMCID: PMC9601531 DOI: 10.3390/genes13101860] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/11/2022] [Accepted: 10/11/2022] [Indexed: 11/04/2022] Open
Abstract
Alopecia areata is a multifactorial autoimmune-based disease with a complex pathogenesis. As in all autoimmune diseases, genetic predisposition is key. The collapse of the immune privilege of the hair follicle leading to scalp loss is a major pathogenic event in alopecia areata. The microbiota considered a bacterial ecosystem located in a specific area of the human body could somehow influence the pathogenesis of alopecia areata, as it occurs in other autoimmune diseases. Moreover, the Next Generation Sequencing of the 16S rRNA bacterial gene and the metagenomic methodology have provided an excellent characterization of the microbiota. The aim of this narrative review is to examine the published literature on the cutaneous and intestinal microbiota in alopecia areata to be able to establish a pathogenic link. In this review, we summarize the influence of the microbiota on the development of alopecia areata. We first introduce the general pathogenic mechanisms that cause alopecia areata to understand the influence that the microbiota may exert and then we summarize the studies that have been carried out on what type of gut and skin microbiota is found in patients with this disease.
Collapse
Affiliation(s)
- Pedro Sánchez-Pellicer
- MiBioPath Research Group, Department of Clinical Medicine, Health Sciences Faculty, Catholic University of Murcia, Campus de los Jerónimos 135, 30107 Murcia, Spain
| | - Laura Navarro-Moratalla
- MiBioPath Research Group, Department of Clinical Medicine, Health Sciences Faculty, Catholic University of Murcia, Campus de los Jerónimos 135, 30107 Murcia, Spain
| | - Eva Núñez-Delegido
- MiBioPath Research Group, Department of Clinical Medicine, Health Sciences Faculty, Catholic University of Murcia, Campus de los Jerónimos 135, 30107 Murcia, Spain
| | - Juan Agüera-Santos
- MiBioPath Research Group, Department of Clinical Medicine, Health Sciences Faculty, Catholic University of Murcia, Campus de los Jerónimos 135, 30107 Murcia, Spain
| | - Vicente Navarro-López
- MiBioPath Research Group, Department of Clinical Medicine, Health Sciences Faculty, Catholic University of Murcia, Campus de los Jerónimos 135, 30107 Murcia, Spain
- Infectious Diseases Unit, University Hospital of Vinalopó-Fisabio, Carrer Tonico Sansano Mora 14, 03293 Elche, Spain
- Correspondence:
| |
Collapse
|
9
|
Li Q, Yang M, Chen K, Zhou S, Zhou S, Wu H. Tight correlation of 5-hydroxymethylcytosine expression with the scarring damage of discoid lupus erythematosus. Lupus 2022; 31:1306-1316. [PMID: 35817588 DOI: 10.1177/09612033221114761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Cutaneous lupus erythematosus (CLE) is a heterogenous skin disease. The two most common subtypes are discoid LE (DLE) characterized by scarring skin damage and acute CLE (ACLE) presenting with transiently reversible skin lesions. It remains unknown what causes the difference of skin lesions. Studies have shown the existence of tissue-specific 5-Hydroxymethylcytosine (5 hmC)-modified regions in human tissues, which may affect the tissue-related diseases. Here, we aim to assess the expression of 5 hmc in DLE and ACLE lesions and explore the relationship of 5 hmc with scarring damage in DLE. METHODS 84 CLE samples were included in the study. We evaluated the skin damage score and reviewed the histopathologic sections. Immunohistochemical staining was performed to detect the expression of 5 hmc in the appendage and periappendageal inflammatory cells. The 5 hmc expression in periappendageal lymphocytic cells was investigated by multi-spectrum immunohistochemistry staining. RESULTS Scarring/atrophy was the most significant damage in differentiating the DLE from ACLE. Perifollicular inflammatory infiltration was present in all patients with DLE scarring alopecia (DLESA). The 5 hmc expression in the appendage and periappendageal inflammatory cells was significantxly increased in DLESA than ACLE. Similar expression pattern was seen in the staining of IFN-alpha/beta Receptor (IFNAR). The expression of 5 hmc in the appendage was positively correlated with that in the periappendageal inflammatory cells. There was an increased 5 hmc expression in lymphocytes cluster around hair follicle consisting of CD4+ cells, CD8+ cells, and CD19+ cells in DLESA lesions. CONCLUSION These data demonstrate a close association of the expression pattern of 5 hmc with the histopathological characteristic distribution, and with the type I interferons (IFNs) signals in DLESA, supporting the importance of 5 hmc in the amplification of appendage damage and periappendageal inflammation, thereby offering a novel insight into the scarring damage of DLE and the heterogeneity of CLE skin lesions.
Collapse
Affiliation(s)
- Qianwen Li
- Department of Dermatology, 70566The Second Xiangya Hospital of Central South University, Changsha, China.,12570The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
| | - Ming Yang
- Department of Dermatology, 70566The Second Xiangya Hospital of Central South University, Changsha, China
| | - Kaili Chen
- Department of Dermatology, 70566The Second Xiangya Hospital of Central South University, Changsha, China.,12570The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
| | - Suqing Zhou
- Department of Dermatology, 70566The Second Xiangya Hospital of Central South University, Changsha, China.,12570The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
| | - Shengnan Zhou
- Department of Dermatology, 70566The Second Xiangya Hospital of Central South University, Changsha, China.,12570The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
| | - Haijing Wu
- Department of Dermatology, 70566The Second Xiangya Hospital of Central South University, Changsha, China.,12570The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
| |
Collapse
|
10
|
Castillo R, Albayda J. Refractory alopecia universalis associated with dermatomyositis successfully treated with tofacitinib. Mod Rheumatol Case Rep 2022; 6:199-202. [PMID: 35253877 PMCID: PMC11132691 DOI: 10.1093/mrcr/rxac012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/26/2021] [Accepted: 02/11/2022] [Indexed: 11/13/2022]
Abstract
Dermatomyositis (DM) and alopecia areata are two diseases characterised by aberrant interferon signalling. While patchy alopecia of the scalp is a known feature of DM, alopecia universalis, which involves hair loss over the entire body, has rarely been reported in conjunction with DM. Herein, we report the case of a 30-year-old female with DM who developed refractory cutaneous disease and alopecia universalis that were successfully treated with tofacitinib. This could suggest that concomitant severe alopecia and refractory cutaneous DM may reflect a strong baseline interferon gene signature that may predict responsiveness to janus kinase inhibitors.
Collapse
Affiliation(s)
- Rochelle Castillo
- Department of Medicine, Division of Rheumatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Jemima Albayda
- Department of Medicine, Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
11
|
Connell SJ, Jabbari A. The current state of knowledge of the immune ecosystem in alopecia areata. Autoimmun Rev 2022; 21:103061. [PMID: 35151885 PMCID: PMC9018517 DOI: 10.1016/j.autrev.2022.103061] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 02/09/2022] [Indexed: 12/18/2022]
Abstract
Alopecia areata (AA) is an autoimmune disease that affects approximately 2% of the general population. Patients with AA most commonly present with one or more patches of hair loss on the scalp in defined circular areas. A fraction of patients progress to more severe forms of the disease, in some cases with involvement of all body surfaces. The healthy anagen stage hair follicle is considered an immune privileged site, described as an environment that suppresses inflammatory immune responses. However, in AA, this immune privileged state collapses and marks the hair follicle as a target for the immune system, resulting in peri- and intrafollicular infiltration by lymphocytes. The complexity of the inflammatory ecosystem of the immune response to the hair follicle, and the relationships between the cellular and soluble participants, in AA remains incompletely understood. Many studies have demonstrated the presence of various immune cells around diseased hair follicles; however, often little is known about their respective contributions to AA pathogenesis. Furthering our understanding of the mechanisms of disease in AA is essential for the novel identification of targeted therapeutics that are efficacious and have few unintended effects.
Collapse
|
12
|
Suchonwanit P, Kositkuljorn C, Pomsoong C. Alopecia Areata: An Autoimmune Disease of Multiple Players. Immunotargets Ther 2021; 10:299-312. [PMID: 34350136 PMCID: PMC8328385 DOI: 10.2147/itt.s266409] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/17/2021] [Indexed: 12/12/2022] Open
Abstract
Alopecia areata (AA) is an autoimmune disease of the hair follicles. It is characterized by a well-defined non-scarring alopecic patch or patches that may extend to the entire scalp or lead to total body hair loss. Due to its unpredictable clinical course, AA causes substantial psychological harm. Despite the high prevalence of this disease and extensive research, its exact pathomechanism is unclear, and current treatments have a high relapse rate that has deemed AA incurable. Over the past few decades, researchers have investigated multiple potential factors that may help alleviate its pathogenesis and provide effective treatment. Given its complex immunopathogenesis, AA is considered an autoimmune disease with multiple factors. This review gathers current evidence that emphasizes molecular mechanisms, possible causative etiologies, and targeted immunotherapies for AA. Understanding its underlying mechanisms may shed light on new strategies to effectively manage AA in the future.
Collapse
Affiliation(s)
- Poonkiat Suchonwanit
- Division of Dermatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Chaninan Kositkuljorn
- Division of Dermatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Cherrin Pomsoong
- Division of Dermatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
13
|
Hashimoto K, Yamada Y, Sekiguchi K, Mori S, Matsumoto T. NLRP3 inflammasome activation contributes to development of alopecia areata in C3H/HeJ mice. Exp Dermatol 2021; 31:133-142. [PMID: 34270818 PMCID: PMC9291919 DOI: 10.1111/exd.14432] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/29/2021] [Accepted: 07/14/2021] [Indexed: 12/31/2022]
Abstract
Alopecia areata (AA) is an autoimmune non‐scarring hair loss disease. Recently, several reports have suggested that innate immune systems such as interferon‐α (IFN‐α)‐producing plasmacytoid dendritic cells and NOD‐like receptor family pyrin domain‐containing protein 3 (NLRP3) inflammasomes play a role in the pathogenesis of AA. However, critical studies about their involvement in the initiation of AA have not yet been reported. Therefore, we investigated the expression of innate immune cytokines in serum and skin, and examined the effect of a selective NLRP3 inhibitor, MCC950, on AA in C3H/HeJ mice, induced by transferring cultured skin‐draining lymph node cells. IFN‐α production was upregulated in lesions of AA‐affected mice, and interleukin‐1β in serum and skin was highly expressed before onset as well as postonset. Furthermore, MCC950 treatment prevented AA development and promoted hair growth in AA mouse models by reducing NLRP3 signalling and Th1/Tc1 chemokines and cytokines in the skin. These results suggest that NLRP3 inflammasome contributes to AA onset and chronicity, and NLRP3 inhibitor may be a potential therapeutic agent for AA.
Collapse
Affiliation(s)
| | | | | | - Sachi Mori
- Kyoto R&D Center, Maruho Co., Ltd., Kyoto, Japan
| | | |
Collapse
|
14
|
Karnell JL, Wu Y, Mittereder N, Smith MA, Gunsior M, Yan L, Casey KA, Henault J, Riggs JM, Nicholson SM, Sanjuan MA, Vousden KA, Werth VP, Drappa J, Illei GG, Rees WA, Ratchford JN. Depleting plasmacytoid dendritic cells reduces local type I interferon responses and disease activity in patients with cutaneous lupus. Sci Transl Med 2021; 13:13/595/eabf8442. [PMID: 34039741 DOI: 10.1126/scitranslmed.abf8442] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 04/12/2021] [Indexed: 12/22/2022]
Abstract
Plasmacytoid dendritic cells (pDCs) not only are specialized in their capacity to secrete large amounts of type I interferon (IFN) but also serve to enable both innate and adaptive immune responses through expression of additional proinflammatory cytokines, chemokines, and costimulatory molecules. Persistent activation of pDCs has been demonstrated in a number of autoimmune diseases. To evaluate the potential benefit of depleting pDCs in autoimmunity, a monoclonal antibody targeting the pDC-specific marker immunoglobulin-like transcript 7 was generated. This antibody, known as VIB7734, which was engineered for enhanced effector function, mediated rapid and potent depletion of pDCs through antibody-dependent cellular cytotoxicity. In cynomolgus monkeys, treatment with VIB7734 reduced pDCs in blood below the lower limit of normal by day 1 after the first dose. In two phase 1 studies in patients with autoimmune diseases, VIB7734 demonstrated an acceptable safety profile, comparable to that of placebo. In individuals with cutaneous lupus, VIB7734 profoundly reduced both circulating and tissue-resident pDCs, with a 97.6% median reduction in skin pDCs at study day 85 in VIB7734-treated participants. Reductions in pDCs in the skin correlated with a decrease in local type I IFN activity as well as improvements in clinical disease activity. Biomarker analysis suggests that responsiveness to pDC depletion therapy may be greater among individuals with high baseline type I IFN activity, supporting a central role for pDCs in type I IFN production in autoimmunity and further development of VIB7734 in IFN-associated diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Li Yan
- Viela Bio, Gaithersburg, MD 20878, USA
| | | | | | | | | | | | | | - Victoria P Werth
- Department of Dermatology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | |
Collapse
|
15
|
Omar SI, Hamza AM, Eldabah N, Habiba DA. IFN-α and TNF-α serum levels and their association with disease severity in Egyptian children and adults with alopecia areata. Int J Dermatol 2021; 60:1397-1404. [PMID: 34008204 DOI: 10.1111/ijd.15658] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 04/09/2021] [Accepted: 04/20/2021] [Indexed: 01/06/2023]
Abstract
BACKGROUND Alopecia areata (AA) is an autoimmune skin disease characterized by abnormal levels of several cytokines, such as interferon alpha (IFN-α) and tumor necrosis factor-alpha (TNF-α), which are T-helper type 1 cytokines that have important roles in the pathogenesis of AA. The aim of our study was to correlate circulating IFN-α and TNF-α levels with disease severity, activity, and clinical type in patients with AA and to evaluate the relationship between the two cytokines. METHODS We investigated serum IFN-α and TNF-α levels in 72 patients with AA (35 children and 35 adults) and 75 healthy control individuals (34 children and 41 adults) using the enzyme-linked immunosorbent assay (ELISA) technique. We evaluated AA severity using the Severity of Alopecia Tool (SALT) and determined the activity based on dermoscopic criteria of disease activity. RESULTS Serum IFN-α and TNF-α concentrations were significantly higher in the patients than in the controls. There was a significant positive correlation between serum IFN-α and TNF-α levels in all patients with alopecia areata, as well as between serum TNF-α levels and disease severity in all patients and in children. CONCLUSIONS Our results support the association between IFN-α and TNF-α levels and AA and suggest that TNF-α might be related to disease severity.
Collapse
Affiliation(s)
- Shaimaa I Omar
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Ashraf M Hamza
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Nermeen Eldabah
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Doaa A Habiba
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
16
|
Dai Z, Chen J, Chang Y, Christiano AM. Selective inhibition of JAK3 signaling is sufficient to reverse alopecia areata. JCI Insight 2021; 6:142205. [PMID: 33830087 PMCID: PMC8119218 DOI: 10.1172/jci.insight.142205] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 02/25/2021] [Indexed: 12/19/2022] Open
Abstract
The Janus kinase/signal transducers and activators of transcription (JAK/STAT) are key intracellular mediators in the signal transduction of many cytokines and growth factors. Common γ chain cytokines and interferon-γ that use the JAK/STAT pathway to induce biological responses have been implicated in the pathogenesis of alopecia areata (AA), a T cell-mediated autoimmune disease of the hair follicle. We previously showed that therapeutic targeting of JAK/STAT pathways using the first-generation JAK1/2 inhibitor, ruxolitinib, and the pan-JAK inhibitor, tofacitinib, was highly effective in the treatment of human AA, as well as prevention and reversal of AA in the C3H/HeJ mouse model. To better define the role of individual JAKs in the pathogenesis of AA, in this study, we tested and compared the efficacy of several next-generation JAK-selective inhibitors in the C3H/HeJ mouse model of AA, using both systemic and topical delivery. We found that JAK1-selective inhibitors as well as JAK3-selective inhibitors robustly induced hair regrowth and decreased AA-associated inflammation, whereas several JAK2-selective inhibitors failed to restore hair growth in treated C3H/HeJ mice with AA. Unlike JAK1, which is broadly expressed in many tissues, JAK3 expression is largely restricted to hematopoietic cells. Our study demonstrates inhibiting JAK3 signaling is sufficient to prevent and reverse disease in the preclinical model of AA.
Collapse
Affiliation(s)
| | | | | | - Angela M. Christiano
- Department of Dermatology and
- Department of Genetics and Development, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
17
|
Abstract
Alopecia areata is a condition that affects hair follicles and leads to hair loss ranging from small well-defined patches to complete loss of all body hair. Despite its high incidence, the pathobiology is not fully understood, and no single concept could be universally accepted. Alopecia areata is mostly considered to be an autoimmune disease, in which the collapse of hair follicle immune privilege plays a key role. Higher incidence rate in the female population and increased overall risk of other autoimmune disorders militate in favor of autoimmune hypothesis. Antibodies against multiple components of hair follicles almost exclusively attack in anagen phase, where melanogenesis takes place. It suggests involvement of melanogenesis-associated autoantigens as a target epitope. Some investigators believed that alopecia areata is not a truly autoimmune disease but is only ‘consistent with’ autoimmune mechanisms. High frequency of a positive family history up to 42% may reflects the contribution of heredity factors. In addition, no specific target autoantigen has been identified so far, and autoantibodies to hair follicle-associated antigens are detectable in normal individuals.
Collapse
|
18
|
Ito T, Kageyama R, Nakazawa S, Honda T. Understanding the significance of cytokines and chemokines in the pathogenesis of alopecia areata. Exp Dermatol 2020; 29:726-732. [DOI: 10.1111/exd.14129] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 05/22/2020] [Accepted: 06/05/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Taisuke Ito
- Department of Dermatology Hamamatsu University School of Medicine Hamamatsu Japan
| | - Reiko Kageyama
- Department of Dermatology Hamamatsu University School of Medicine Hamamatsu Japan
| | - Shinsuke Nakazawa
- Department of Dermatology Hamamatsu University School of Medicine Hamamatsu Japan
| | - Tetsuya Honda
- Department of Dermatology Hamamatsu University School of Medicine Hamamatsu Japan
| |
Collapse
|
19
|
Mitra D, Chopra A, Saraswat N, Mitra B, Talukdar K, Agarwal R. Biologics in Dermatology: Off-Label Indications. Indian Dermatol Online J 2020; 11:319-327. [PMID: 32695686 PMCID: PMC7367577 DOI: 10.4103/idoj.idoj_407_18] [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: 11/25/2018] [Revised: 06/05/2019] [Accepted: 06/10/2019] [Indexed: 11/04/2022] Open
Abstract
Skin and subcutaneous diseases affect millions of people worldwide, causing significant morbidity. Biologics are becoming increasingly useful for the treatment of many skin diseases, particularly as alternatives for patients who have failed to tolerate or respond to conventional systemic therapies. Biological therapies provide a targeted approach to treatment through interaction with specific components of the underlying immune and inflammatory disease processes. Advances in the understanding of disease pathophysiology for inflammatory skin diseases and in drug development have ushered in biologic therapies in dermatology. Biologic therapies are molecules that target specific proteins implicated in immune-mediated disease. This review article highlights the increasing evidence base for biologics in dermatology for off-label use.
Collapse
Affiliation(s)
- Debdeep Mitra
- Department of Dermatology, Base Hospital Delhi Cantt, New Delhi, India
| | - Ajay Chopra
- Department of Dermatology, Base Hospital Delhi Cantt, New Delhi, India
| | - Neerja Saraswat
- Department of Dermatology, Base Hospital Delhi Cantt, New Delhi, India
| | - Barnali Mitra
- Department of Pediatrics, Base Hospital Delhi Cantt, New Delhi, India
| | - Krishna Talukdar
- Department of Dermatology, Jorhat Medical College and Hospital, Jorhat, Assam, India
| | - Reetu Agarwal
- Department of Dermatology, Base Hospital Delhi Cantt, New Delhi, India
| |
Collapse
|
20
|
Kang H, Wu W, Yu M, Shapiro J, McElwee KJ. Increased expression of TLR7 and TLR9 in alopecia areata. Exp Dermatol 2019; 29:254-258. [DOI: 10.1111/exd.14043] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 08/20/2019] [Accepted: 09/21/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Hoon Kang
- Department of Dermatology College of Medicine The Catholic University of Korea Seoul Korea
| | - Wen‐Yu Wu
- Department of Dermatology Huashan Hospital Fudan University Shanghai China
| | - Mei Yu
- Department of Dermatology and Skin Science University of British Columbia Vancouver BC Canada
| | - Jerry Shapiro
- The Ronald O. Perelman Department of Dermatology New York University School of Medicine New York NY USA
| | | |
Collapse
|
21
|
Howell MD, Kuo FI, Smith PA. Targeting the Janus Kinase Family in Autoimmune Skin Diseases. Front Immunol 2019; 10:2342. [PMID: 31649667 PMCID: PMC6794457 DOI: 10.3389/fimmu.2019.02342] [Citation(s) in RCA: 160] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 09/17/2019] [Indexed: 12/12/2022] Open
Abstract
Autoimmune skin diseases are characterized by significant local and systemic inflammation that is largely mediated by the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway. Advanced understanding of this pathway has led to the development of targeted inhibitors of Janus kinases (JAKinibs). As a class, JAK inhibitors effectively treat a multitude of hematologic and inflammatory diseases. Growing evidence suggests that JAK inhibitors are efficacious in atopic dermatitis, alopecia areata, psoriasis, and vitiligo. Additional evidence suggests that JAK inhibition might be broadly useful in dermatology, with early reports of efficacy in several other conditions. JAK inhibitors can be administered orally or used topically and represent a promising new class of medications. Here we review the evolving data on the role of the JAK-STAT pathway in inflammatory dermatoses and the potential therapeutic benefit of JAK-STAT antagonism.
Collapse
Affiliation(s)
| | - Fiona I Kuo
- Incyte Corporation, Wilmington, DE, United States
| | - Paul A Smith
- Incyte Corporation, Wilmington, DE, United States
| |
Collapse
|
22
|
Richmond JM, Strassner JP, Essien KI, Harris JE. T-cell positioning by chemokines in autoimmune skin diseases. Immunol Rev 2019; 289:186-204. [PMID: 30977191 PMCID: PMC6553463 DOI: 10.1111/imr.12762] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 03/18/2019] [Accepted: 03/21/2019] [Indexed: 12/12/2022]
Abstract
Autoimmune skin diseases are complex processes in which autoreactive cells must navigate through the skin tissue to find their targets. Regulatory T cells in the skin help to mitigate autoimmune inflammation and may in fact be responsible for the patchy nature of these conditions. In this review, we will discuss chemokines that are important for global recruitment of T cell populations to the skin during disease, as well as signals that fine-tune their localization and function. We will describe prototypical disease responses and chemokine families that mediate these responses. Lastly, we will include an overview of chemokine-targeting drugs that have been tested as new treatment strategies for autoimmune skin diseases.
Collapse
Affiliation(s)
- Jillian M Richmond
- Department of Dermatology, UMass Medical School, Worcester, Massachusetts
| | - James P Strassner
- Department of Dermatology, UMass Medical School, Worcester, Massachusetts
| | - Kingsley I Essien
- Department of Dermatology, UMass Medical School, Worcester, Massachusetts
| | - John E Harris
- Department of Dermatology, UMass Medical School, Worcester, Massachusetts
| |
Collapse
|
23
|
Rajabi F, Amoli MM, Robati RM, Almasi-Nasrabadi M, Jabalameli N, Moravvej H. The Association between Genetic Variation in Wnt Transcription FactorTCF7L2(TCF4) and Alopecia Areata. Immunol Invest 2019; 48:555-562. [DOI: 10.1080/08820139.2019.1597109] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Fateme Rajabi
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Network of Dermatology Research (NDR), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahsa M. Amoli
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular -Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza M Robati
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Navid Jabalameli
- Network of Dermatology Research (NDR), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hamideh Moravvej
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Turner CT, Lim D, Granville DJ. Granzyme B in skin inflammation and disease. Matrix Biol 2019; 75-76:126-140. [DOI: 10.1016/j.matbio.2017.12.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/11/2017] [Accepted: 12/12/2017] [Indexed: 01/30/2023]
|
25
|
Simakou T, Butcher JP, Reid S, Henriquez FL. Alopecia areata: A multifactorial autoimmune condition. J Autoimmun 2018; 98:74-85. [PMID: 30558963 DOI: 10.1016/j.jaut.2018.12.001] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 12/04/2018] [Accepted: 12/06/2018] [Indexed: 02/07/2023]
Abstract
Alopecia areata is an autoimmune disease that results in non-scarring hair loss, and it is clinically characterised by small patches of baldness on the scalp and/or around the body. It can later progress to total loss of scalp hair (Alopecia totalis) and/or total loss of all body hair (Alopecia universalis). The rapid rate of hair loss and disfiguration caused by the condition causes anxiety on patients and increases the risks of developing psychological and psychiatric complications. Hair loss in alopecia areata is caused by lymphocytic infiltrations around the hair follicles and IFN-γ. IgG antibodies against the hair follicle cells are also found in alopecia areata sufferers. In addition, the disease coexists with other autoimmune disorders and can come secondary to infections or inflammation. However, despite the growing knowledge about alopecia areata, the aetiology and pathophysiology of disease are not well defined. In this review we discuss various genetic and environmental factors that cause autoimmunity and describe the immune mechanisms that lead to hair loss in alopecia areata patients.
Collapse
Affiliation(s)
- Teontor Simakou
- Institute of Biomedical and Environmental Health Research, School of Health and Life Sciences, University of the West of Scotland, 1 High Street, Paisley, PA1 2BE, UK
| | - John P Butcher
- Department of Life Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow, G4 0BA, UK
| | - Stuart Reid
- SUPA, Department of Biomedical Engineering, University of Strathclyde, UK
| | - Fiona L Henriquez
- Institute of Biomedical and Environmental Health Research, School of Health and Life Sciences, University of the West of Scotland, 1 High Street, Paisley, PA1 2BE, UK.
| |
Collapse
|
26
|
Rajabi F, Drake LA, Senna MM, Rezaei N. Alopecia areata: a review of disease pathogenesis. Br J Dermatol 2018; 179:1033-1048. [PMID: 29791718 DOI: 10.1111/bjd.16808] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Alopecia areata is a disorder that results in nonscarring hair loss. The psychological impact can be significant, leading to feelings of depression and social isolation. Objectives In this article, we seek to review the pathophysiological mechanisms proposed in recent years in a narrative fashion. METHODS We searched MEDLINE and Scopus for articles related to alopecia areata, with a particular emphasis on its pathogenesis. RESULTS The main theory of alopecia areata pathogenesis is that it is an autoimmune phenomenon resulting from a disruption in hair follicle immune privilege. What causes this breakdown is an issue of debate. Some believe that a stressed hair follicle environment triggers antigen presentation, while others blame a dysregulation in the central immune system entangling the follicles. Evidence for the latter theory is provided by animal studies, as well investigations around the AIRE gene. Different immune-cell lines including plasmacytoid dendritic cells, natural killer cells and T cells, along with key molecules such as interferon-γ, interleukin-15, MICA and NKG2D, have been identified as contributing to the autoimmune process. CONCLUSIONS Alopecia areata remains incurable, although it has been studied for years. Available treatment options at best are beneficial for milder cases, and the rate of relapse is high. Understanding the exact mechanisms of hair loss in alopecia areata is therefore of utmost importance to help identify potential therapeutic targets.
Collapse
Affiliation(s)
- F Rajabi
- Department of Dermatology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - L A Drake
- Massachusetts General Hospital, Harvard Medical School, MA, U.S.A
| | - M M Senna
- Massachusetts General Hospital, Harvard Medical School, MA, U.S.A
| | - N Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Sheffield, U.K
| |
Collapse
|
27
|
Wang EHC, Sallee BN, Tejeda CI, Christiano AM. JAK Inhibitors for Treatment of Alopecia Areata. J Invest Dermatol 2018; 138:1911-1916. [PMID: 30057345 DOI: 10.1016/j.jid.2018.05.027] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 05/22/2018] [Accepted: 05/27/2018] [Indexed: 01/25/2023]
Abstract
The advancement of genetic and preclinical studies has uncovered the mechanisms involved in the pathogenesis of alopecia areata (AA). The development of targeted therapies using small molecules blocking specific pathways for the treatment of AA is underway. By repurposing Food and Drug Administration-approved small molecule JAK inhibitors as treatments for AA, it has been demonstrated that JAK inhibitors can effectively reverse hair loss in patients with moderate to severe AA. In this review, we summarize and discuss the current preclinical and clinical studies on JAK inhibitors, as well as the prospects of using JAK inhibitors for the treatment of AA.
Collapse
Affiliation(s)
- Eddy H C Wang
- Department of Dermatology, Columbia University, New York, New York, USA
| | - Brigitte N Sallee
- Department of Dermatology, Columbia University, New York, New York, USA
| | - Christina I Tejeda
- Columbia College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Angela M Christiano
- Department of Dermatology, Columbia University, New York, New York, USA; Genetics and Development, Columbia University, New York, New York, USA.
| |
Collapse
|
28
|
Evaluation of the Relationship between Alopecia Areata and Viral Antigen Exposure. Am J Clin Dermatol 2018; 19:119-126. [PMID: 28801732 DOI: 10.1007/s40257-017-0312-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Alopecia areata (AA) is an autoimmune disease characterized by non-scarring alopecia with T-cell infiltration at the affected hair follicle. OBJECTIVE Our aim was to study the potential link between hepatitis B virus (HBV) antigen exposure and AA. METHODS Two pediatric patients with AA following hepatitis B vaccination were identified in a general dermatology clinic. A bioinformatics analysis and an electronic medical record (EMR) database query were performed at the University of Rochester Medical Center to identify patients with AA, coexisting viral infections, vaccinations, or interferon (IFN) therapy in order to determine if the presence of AA and these conditions was higher than in AA patients without these associated conditions or therapy. RESULTS An increased frequency of AA among those who received the HBV surface protein antigen [odds ratio (OR) 2.7, p < 0.0001] was identified, and an independent analysis revealed an increased frequency of AA in those receiving IFN-β treatment (OR 8.1, p < 0.05). One potential antigenic target identified was SLC45A2, a melanosomal transport protein important in skin and hair pigmentation. The longest potential vaccine peptide fragment match (8-mer) was to a segment of natural killer (NK) cell inhibitory receptors, KIR3DL2 and KIR3DL1. Predictive modeling of major histocompatibility complex (MHC)-peptide binding demonstrated potential binding of this peptide to MHC relevant to AA. LIMITATIONS The results will need to be verified in additional patient databases allowing analysis of temporal relationships, and with molecular experiments of the identified antigens. CONCLUSIONS Our data confirm associations between viral infection and IFN treatment with AA. It establishes that the hepatitis B surface protein antigen has shared epitopes with human killer immunoglobulin-like receptors.
Collapse
|
29
|
Abstract
Few studies have examined the clinical epidemiology of alopecia areata (AA) in regard to patient race, and therefore, any disparities in incidence or prevalence of disease are largely unexplored. We sought to investigate potential racial disparities amongst two large cohorts of women. We conducted a cross-sectional analysis from the Nurses' Health Study (NHS) and Nurses' Health Study II (NHSII), wherein participants self-reported a diagnosis of AA. We determined odds ratios for AA by race in a multivariate analysis. Among 63,960 women from NHS and 88,368 women from NHSII with information on race and diagnosis of AA, we identified 418 and 738 cases of AA, respectively. In NHS, the multivariate-adjusted odds ratio for AA was 2.72 (95% confidence interval 1.61-4.61) amongst black women as compared with white women. In NHSII, the multivariate-adjusted odds ratio was 5.48 (95% confidence interval 4.10-7.32) amongst black as compared with white women. In a secondary analysis designating participants by Hispanic ethnicity, in NHSII the multivariate odds ratio was 1.94 (95% CI 1.24-3.02) in Hispanic compared with non-Hispanic white women. In this study, we found increased odds of AA based on self-reported race in black and Hispanic women as compared with white women. Further studies are needed to explore the mechanism of this racial disparity related to AA.
Collapse
Affiliation(s)
- Jordan M Thompson
- Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Min Kyung Park
- Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Abrar A Qureshi
- Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA; Department of Epidemiology, School of Public Health, Brown University, Providence, Rhode Island, USA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Eunyoung Cho
- Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA; Department of Epidemiology, School of Public Health, Brown University, Providence, Rhode Island, USA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
30
|
Wang ECE, Dai Z, Christiano AM. Novel therapies for alopecia areata: The era of rational drug development. J Allergy Clin Immunol 2017; 141:499-504. [PMID: 29155099 DOI: 10.1016/j.jaci.2017.10.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 09/29/2017] [Accepted: 10/11/2017] [Indexed: 12/19/2022]
Abstract
Treatments for alopecia areata (AA) have evolved over the decades from broad and nonspecific therapies to those that are now more targeted and rationally selected. This was achieved by means of close cooperation and communication between clinicians and basic scientists, which resulted in the elucidation and understanding of the unique pathophysiology of AA. In this review we discuss this evolution and how novel therapies for AA have changed over the decades, what we have in our current arsenal of drugs for this potentially devastating disease, and what the future holds.
Collapse
Affiliation(s)
- Etienne C E Wang
- Department of Dermatology, Columbia University, New York, NY; National Skin Center, Singapore
| | - Zhenpeng Dai
- Department of Dermatology, Columbia University, New York, NY
| | | |
Collapse
|
31
|
Abstract
Alopecia areata is an autoimmune disorder characterized by transient, non-scarring hair loss and preservation of the hair follicle. Hair loss can take many forms ranging from loss in well-defined patches to diffuse or total hair loss, which can affect all hair-bearing sites. Patchy alopecia areata affecting the scalp is the most common type. Alopecia areata affects nearly 2% of the general population at some point during their lifetime. Skin biopsies of affected skin show a lymphocytic infiltrate in and around the bulb or the lower part of the hair follicle in the anagen (hair growth) phase. A breakdown of immune privilege of the hair follicle is thought to be an important driver of alopecia areata. Genetic studies in patients and mouse models have shown that alopecia areata is a complex, polygenic disease. Several genetic susceptibility loci were identified to be associated with signalling pathways that are important to hair follicle cycling and development. Alopecia areata is usually diagnosed based on clinical manifestations, but dermoscopy and histopathology can be helpful. Alopecia areata is difficult to manage medically, but recent advances in understanding the molecular mechanisms have revealed new treatments and the possibility of remission in the near future.
Collapse
Affiliation(s)
- C Herbert Pratt
- Department of Genetic Resource Sciences, The Jackson Laboratory, Bar Harbor, Maine, USA
| | - Lloyd E King
- Department of Dermatology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Angela M Christiano
- Departments of Dermatology and Genetics &Development, Columbia University, New York, New York, USA
| | - John P Sundberg
- Department of Dermatology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Research and Development, The Jackson Laboratory, 600 Main Street, Bar Harbor, Maine 04609-1500, USA
| |
Collapse
|
32
|
Woo YR, Hwang S, Jeong SW, Cho DH, Park HJ. Erythroid Differentiation Regulator 1 as a Novel Biomarker for Hair Loss Disorders. Int J Mol Sci 2017; 18:ijms18020316. [PMID: 28165377 PMCID: PMC5343852 DOI: 10.3390/ijms18020316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/24/2017] [Accepted: 01/27/2017] [Indexed: 11/16/2022] Open
Abstract
Erythroid differentiation regulator 1 (Erdr1) is known to be involved in the inflammatory process via regulating the immune system in many cutaneous disorders, such as psoriasis and rosacea. However, the role of Erdr1 in various hair loss disorders remains unclear. The aim of this study was to investigate the putative role of Erdr1 in alopecias. Skin samples from 21 patients with hair loss disorders and five control subjects were retrieved, in order to assess their expression levels of Erdr1. Results revealed that expression of Erdr1 was significantly downregulated in the epidermis and hair follicles of patients with hair loss disorders, when compared to that in the control group. In particular, the expression of Erdr1 was significantly decreased in patients with alopecia areata. We propose that Erdr1 downregulation might be involved in the pathogenesis of hair loss, and could be considered as a novel biomarker for hair loss disorders.
Collapse
Affiliation(s)
- Yu Ri Woo
- Department of Dermatology, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 07345, Korea.
| | - Sewon Hwang
- Department of Dermatology, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 07345, Korea.
| | - Seo Won Jeong
- Institute of Clinical Medical Research, Yeouido St. Mary's Hostpital, College of Medicine, The Catholic University of Korea, Seoul 07345, Korea.
| | - Dae Ho Cho
- Department of Life Science, Sookmyung Women's University, Seoul 04310, Korea.
| | - Hyun Jeong Park
- Department of Dermatology, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 07345, Korea.
| |
Collapse
|
33
|
Abstract
Alopecia areata (AA) is non-scarring hair loss resulting from an autoimmune disorder. Severity varies from patchy hair loss that often spontaneously resolves to severe and chronic cases that can progress to total loss of scalp and body hair. Many treatments are available; however, the efficacy of these treatments has not been confirmed, especially in severe cases, and relapse rates are high. First-line treatment often includes corticosteroids such as intralesional or topical steroids for mild cases and systemic steroids or topical immunotherapy with diphenylcyclopropenone or squaric acid dibutylester in severe cases. Minoxidil and bimatoprost may also be recommended, usually in combination with another treatment. Ongoing research and new insights into mechanisms have led to proposals of innovative therapies. New directions include biologics targeting immune response as well as lasers and autologous platelet-rich plasma therapy. Preliminary data are encouraging, and it is hoped this research will translate into new options for the treatment of AA in the near future.
Collapse
|
34
|
Saadeh D, Kurban M, Abbas O. Update on the role of plasmacytoid dendritic cells in inflammatory/autoimmune skin diseases. Exp Dermatol 2016; 25:415-21. [PMID: 26837058 DOI: 10.1111/exd.12957] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2016] [Indexed: 12/28/2022]
Abstract
Plasmacytoid dendritic cells (pDCs) represent a specialized dendritic cell population that exhibit plasma cell morphology, express CD4, CD123, blood-derived dendritic cell antigen-2 (BDCA-2) and Toll-like receptor (TLR)7 and TLR9 within endosomal compartments. When activated, pDCs are capable of producing large quantities of type I IFNs (mainly IFN-α/β), which provide antiviral resistance and link the innate and adaptive immunity. While generally lacking from normal skin, pDCs infiltrate the skin and appear to be involved in the pathogenesis of several inflammatory, infectious (especially viral) and neoplastic entities. In recent years, pDC role in inflammatory/autoimmune skin conditions has been extensively studied. Unlike type I IFN-mediated protective immunity that pDCs provide at the level of the skin by regulated sensing of microbial or self-nucleic acids upon skin damage, excessive sensing may elicit IFN-driven inflammatory/autoimmune diseases. In this review, focus will be on the role of pDCs in cutaneous inflammatory/autoimmune dermatoses.
Collapse
Affiliation(s)
- Dana Saadeh
- Dermatology Department, American University of Beirut Medical Center, Beirut, Lebanon
| | - Mazen Kurban
- Dermatology Department, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ossama Abbas
- Dermatology Department, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
35
|
Shen X, Nair B, Mahajan SD, Jiang X, Li J, Shen S, Tu C, Hsiao CB, Schwartz SA, Qu J. New Insights into the Disease Progression Control Mechanisms by Comparing Long-Term-Nonprogressors versus Normal-Progressors among HIV-1-Positive Patients Using an Ion Current-Based MS1 Proteomic Profiling. J Proteome Res 2015; 14:5225-39. [PMID: 26484939 DOI: 10.1021/acs.jproteome.5b00621] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
For decades, epidemiological studies have found significant differences in the susceptibility to disease progression among HIV-carrying patients. One unique group of HIV-1-positive patients, the long-term-nonprogressors (LTNP), exhibits far superior ability in virus control compared with normal-progressors (NP), which proceed to Acquired Immune Deficiency Syndrome (AIDS) much more rapidly. Nonetheless, elucidation of the underlying mechanisms of virus control in LTNP is highly valuable in disease management and treatment but remains poorly understood. Peripheral blood mononuclear cells (PBMC) have been known to play important roles in innate immune responses and thereby would be of great interest for the investigation of the mechanisms of virus defense in LTNP. Here, we described the first comparative proteome analysis of PBMC from LTNP (n = 10) and NP (n = 10) patients using a reproducible ion-current-based MS1 approach, which includes efficient and reproducible sample preparation and chromatographic separation followed by an optimized pipeline for protein identification and quantification. This strategy enables analysis of many biological samples in one set with high quantitative precision and extremely low missing data. In total, 925 unique proteins were quantified under stringent criteria without missing value in any of the 20 subjects, and 87 proteins showed altered expressions between the two patient groups. These proteins are implicated in key processes such as cytoskeleton organization, defense response, apoptosis regulation, intracellular transport, etc., which provided novel insights into the control of disease progressions in LTNP versus NP, and the expression and phosphorylation states of key regulators were further validated by immunoassay. For instance, (1) SAMH1, a potent and "hot" molecule facilitating HIV-1 defense, was for the first time found elevated in LTNP compared with NP or healthy controls; elevated proteins from IFN-α response pathway may also contribute to viral control in LTNP; (2) decreased proapoptotic protein ASC along with the elevation of antiapoptotic proteins may contribute to the less apoptotic profile in PBMC of LTNP; and (3) elevated actin polymerization and less microtubule assembly that impede viral protein transport were first observed in LTNP. These results not only enhanced the understanding of the mechanisms for nonprogression of LTNP, but also may afford highly valuable clues to direct therapeutic efforts. Moreover, this work also demonstrated the ion-current-based MS1 approach as a reliable tool for large-scale clinical research.
Collapse
Affiliation(s)
- Xiaomeng Shen
- The State of New York Center for Excellence in Bioinformatics and Life Science, 701 Ellicott Street, Buffalo, New York 14203, United States
| | | | | | - Xiaosheng Jiang
- The State of New York Center for Excellence in Bioinformatics and Life Science, 701 Ellicott Street, Buffalo, New York 14203, United States
| | - Jun Li
- The State of New York Center for Excellence in Bioinformatics and Life Science, 701 Ellicott Street, Buffalo, New York 14203, United States
| | - Shichen Shen
- The State of New York Center for Excellence in Bioinformatics and Life Science, 701 Ellicott Street, Buffalo, New York 14203, United States
| | - Chengjian Tu
- The State of New York Center for Excellence in Bioinformatics and Life Science, 701 Ellicott Street, Buffalo, New York 14203, United States
| | - Chiu-Bin Hsiao
- Infectious Disease Division, Department of Medicine, Allegheny General Hospital , Pittsburgh, Pennsylvania 15212, United States
| | | | - Jun Qu
- The State of New York Center for Excellence in Bioinformatics and Life Science, 701 Ellicott Street, Buffalo, New York 14203, United States
| |
Collapse
|
36
|
|
37
|
Jabbari A, Dai Z, Xing L, Cerise JE, Ramot Y, Berkun Y, Sanchez GAM, Goldbach-Mansky R, Christiano AM, Clynes R, Zlotogorski A. Reversal of Alopecia Areata Following Treatment With the JAK1/2 Inhibitor Baricitinib. EBioMedicine 2015; 2:351-5. [PMID: 26137574 PMCID: PMC4486197 DOI: 10.1016/j.ebiom.2015.02.015] [Citation(s) in RCA: 174] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 02/20/2015] [Accepted: 02/24/2015] [Indexed: 12/24/2022] Open
Abstract
Background Alopecia areata (AA) is an autoimmune disease resulting in hair loss with devastating psychosocial consequences. Despite its high prevalence, there are no FDA-approved treatments for AA. Prior studies have identified a prominent interferon signature in AA, which signals through JAK molecules. Methods A patient with AA was enrolled in a clinical trial to examine the efficacy of baricitinib, a JAK1/2 inhibitor, to treat concomitant CANDLE syndrome. In vivo, preclinical studies were conducted using the C3H/HeJ AA mouse model to assess the mechanism of clinical improvement by baricitinib. Findings The patient exhibited a striking improvement of his AA on baricitinib over several months. In vivo studies using the C3H/HeJ mouse model demonstrated a strong correlation between resolution of the interferon signature and clinical improvement during baricitinib treatment. Interpretation Baricitinib may be an effective treatment for AA and warrants further investigation in clinical trials.
Collapse
Affiliation(s)
- Ali Jabbari
- Department of Dermatology, Columbia University, New York, NY, USA
| | - Zhenpeng Dai
- Department of Dermatology, Columbia University, New York, NY, USA
| | - Luzhou Xing
- Department of Pathology, Columbia University, New York, NY, USA
| | - Jane E Cerise
- Department of Dermatology, Columbia University, New York, NY, USA
| | - Yuval Ramot
- Department of Dermatology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Yackov Berkun
- Department of Pediatrics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | | | - Angela M Christiano
- Department of Dermatology, Columbia University, New York, NY, USA ; Department of Genetics & Development, Columbia University, New York, NY, USA
| | - Raphael Clynes
- Department of Dermatology, Columbia University, New York, NY, USA ; Department of Pathology, Columbia University, New York, NY, USA ; Department of Medicine, Columbia University, New York, NY, USA
| | - Abraham Zlotogorski
- Department of Dermatology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
38
|
Abou Rahal J, Kurban M, Kibbi AG, Abbas O. Plasmacytoid dendritic cells in alopecia areata: missing link? J Eur Acad Dermatol Venereol 2014; 30:119-23. [DOI: 10.1111/jdv.12932] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 11/20/2014] [Indexed: 11/30/2022]
Affiliation(s)
- J. Abou Rahal
- Dermatology Department; American University of Beirut Medical Center; Beirut Lebanon
| | - M. Kurban
- Dermatology Department; American University of Beirut Medical Center; Beirut Lebanon
| | - A.-G. Kibbi
- Dermatology Department; American University of Beirut Medical Center; Beirut Lebanon
| | - O. Abbas
- Dermatology Department; American University of Beirut Medical Center; Beirut Lebanon
| |
Collapse
|
39
|
Ito T, Tokura Y. The role of cytokines and chemokines in the T-cell-mediated autoimmune process in alopecia areata. Exp Dermatol 2014; 23:787-91. [DOI: 10.1111/exd.12489] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2014] [Indexed: 12/12/2022]
Affiliation(s)
- Taisuke Ito
- Department of Dermatology; Hamamatsu University School of Medicine; Hamamatsu Japan
| | - Yoshiki Tokura
- Department of Dermatology; Hamamatsu University School of Medicine; Hamamatsu Japan
| |
Collapse
|
40
|
McElwee KJ, Gilhar A, Tobin DJ, Ramot Y, Sundberg JP, Nakamura M, Bertolini M, Inui S, Tokura Y, Jr LEK, Duque-Estrada B, Tosti A, Keren A, Itami S, Shoenfeld Y, Zlotogorski A, Paus R. What causes alopecia areata? Exp Dermatol 2013; 22:609-26. [PMID: 23947678 PMCID: PMC4094373 DOI: 10.1111/exd.12209] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The pathobiology of alopecia areata (AA), one of the most frequent autoimmune diseases and a major unsolved clinical problem, has intrigued dermatologists, hair biologists and immunologists for decades. Simultaneously, both affected patients and the physicians who take care of them are increasingly frustrated that there is still no fully satisfactory treatment. Much of this frustration results from the fact that the pathobiology of AA remains unclear, and no single AA pathogenesis concept can claim to be universally accepted. In fact, some investigators still harbour doubts whether this even is an autoimmune disease, and the relative importance of CD8(+) T cells, CD4(+) T cells and NKGD2(+) NK or NKT cells and the exact role of genetic factors in AA pathogenesis remain bones of contention. Also, is AA one disease, a spectrum of distinct disease entities or only a response pattern of normal hair follicles to immunologically mediated damage? During the past decade, substantial progress has been made in basic AA-related research, in the development of new models for translationally relevant AA research and in the identification of new therapeutic agents and targets for future AA management. This calls for a re-evaluation and public debate of currently prevalent AA pathobiology concepts. The present Controversies feature takes on this challenge, hoping to attract more skin biologists, immunologists and professional autoimmunity experts to this biologically fascinating and clinically important model disease.
Collapse
Affiliation(s)
- K. J. McElwee
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC, Canada
| | - A. Gilhar
- Laboratory for Skin, Research, Rappaport Faculty of Medicine, Technion–Israel Institute of Technology, Haifa, Israel Marta Bertolini
| | - D. J. Tobin
- Centre for Skin Sciences, School of Life Sciences, University of Bradford, Bradford, West Yorkshire, BD7 1DP, UK
| | - Y. Ramot
- Department of Dermatology, Hadassah- Hebrew University Medical Center, Jerusalem 91120, Israel
| | - J. P. Sundberg
- The Jackson Laboratory, Bar Harbor, ME, USA; Division of Dermatology, Skin Disease Research Center, Vanderbilt University, Nashville, TN, USA
| | - M. Nakamura
- Department of Dermatology, University of Occupational and Environmental Health, Kitakyushu, Japan Yoshiki Tokura
| | - M. Bertolini
- Department of Dermatology, University of Lübeck, Germany Yehuda Shoenfeld
| | - S. Inui
- Department of Regenerative Dermatology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Y. Tokura
- Department of Dermatology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - L. E. King Jr
- The Jackson Laboratory, Bar Harbor, ME, USA; Division of Dermatology, Skin Disease Research Center, Vanderbilt University, Nashville, TN, USA
| | - B. Duque-Estrada
- Instituto de Dermatologia Prof. Rubem David Azulay, Rio de Janeiro, Brazil Antonella Tosti
| | - A Tosti
- Department of Dermatology, University of Miami, Miami, FL, USA
| | - A. Keren
- Laboratory for Skin, Research, Rappaport Faculty of Medicine, Technion–Israel Institute of Technology, Haifa, Israel Marta Bertolini
| | - S. Itami
- Department of Regenerative Dermatology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Y. Shoenfeld
- Center for Autoimmune Diseases, Sheba Medical Center, Tel Hashomer and Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Israel
| | - A. Zlotogorski
- Department of Dermatology, Hadassah- Hebrew University Medical Center, Jerusalem 91120, Israel
| | - R. Paus
- Department of Dermatology, University of Lübeck, Germany; Institute of Inflammation and Repair, University of Manchester, Manchester, UK ,
| |
Collapse
|
41
|
Malard F, Bossard C, Brissot E, Chevallier P, Guillaume T, Delaunay J, Mosnier JF, Moreau P, Grégoire M, Gaugler B, Mohty M. Increased plasmacytoid dendritic cells and RORγt-expressing immune effectors in cutaneous acute graft-versus-host disease. J Leukoc Biol 2013; 94:1337-43. [PMID: 23990625 DOI: 10.1189/jlb.0513295] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The role of PDCs and Th17 cells is not well understood in the pathogenesis of aGVHD. We evaluated PDC and Th17 cells in skin biopsies of 38 patients at diagnosis of aGVHD. The biopsies were tested by immunohistochemistry for the expression of BDCA2, a typical marker of PDCs. We found an increase of BDCA2(+) cells in the skin of the patients with aGVHD. Moreover, we observed a strong expression of the type I IFN-inducible protein Mx1 in the skin of the patients with aGVHD, compared with that of those without it, suggesting that PDCs produce type I IFN. We also analyzed the expression of two Th17 surface markers-CD161 and CCR6-and RORγt, the key transcription factor that orchestrates the differentiation of Th17 cells. Significantly higher numbers of RORγt(+), CD161(+), and CCR6(+) cells were counted in the skin of the patients with aGVHD than in the skin of those who underwent allo-SCT and in whom aGVHD did not develop. This study provides evidence for a role of Th17-mediated responses and a potential new pathophysiological link between PDCs and Th17 in human cutaneous aGVHD.
Collapse
Affiliation(s)
- Florent Malard
- 3.CHU Hôtel-Dieu, Université de Nantes and INSERM U892, Place Alexis Ricordeau, F-44093 Nantes, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Nagao K, Kobayashi T, Moro K, Ohyama M, Adachi T, Kitashima DY, Ueha S, Horiuchi K, Tanizaki H, Kabashima K, Kubo A, Cho YH, Clausen BE, Matsushima K, Suematsu M, Furtado GC, Lira SA, Farber JM, Udey MC, Amagai M. Stress-induced production of chemokines by hair follicles regulates the trafficking of dendritic cells in skin. Nat Immunol 2012; 13:744-52. [PMID: 22729248 DOI: 10.1038/ni.2353] [Citation(s) in RCA: 256] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 05/30/2012] [Indexed: 01/14/2023]
Abstract
Langerhans cells (LCs) are epidermal dendritic cells with incompletely understood origins that associate with hair follicles for unknown reasons. Here we show that in response to external stress, mouse hair follicles recruited Gr-1(hi) monocyte-derived precursors of LCs whose epidermal entry was dependent on the chemokine receptors CCR2 and CCR6, whereas the chemokine receptor CCR8 inhibited the recruitment of LCs. Distinct hair-follicle regions had differences in their expression of ligands for CCR2 and CCR6. The isthmus expressed the chemokine CCL2; the infundibulum expressed the chemokine CCL20; and keratinocytes in the bulge produced the chemokine CCL8, which is the ligand for CCR8. Thus, distinct hair-follicle keratinocyte subpopulations promoted or inhibited repopulation with LCs via differences in chemokine production, a feature also noted in humans. Pre-LCs failed to enter hairless skin in mice or humans, which establishes hair follicles as portals for LCs.
Collapse
Affiliation(s)
- Keisuke Nagao
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Ghoreishi M, Vera Kellet C, Dutz JP. Type 1 IFN-induced protein MxA and plasmacytoid dendritic cells in lesions of morphea. Exp Dermatol 2012; 21:417-9. [DOI: 10.1111/j.1600-0625.2012.01475.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
44
|
McPhee CG, Duncan FJ, Silva KA, King LE, Hogenesch H, Roopenian DC, Everts HB, Sundberg JP. Increased expression of Cxcr3 and its ligands, Cxcl9 and Cxcl10, during the development of alopecia areata in the mouse. J Invest Dermatol 2012; 132:1736-8. [PMID: 22358057 PMCID: PMC3352958 DOI: 10.1038/jid.2012.17] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
45
|
Wong D, Kea B, Pesich R, Higgs BW, Zhu W, Brown P, Yao Y, Fiorentino D. Interferon and biologic signatures in dermatomyositis skin: specificity and heterogeneity across diseases. PLoS One 2012; 7:e29161. [PMID: 22235269 PMCID: PMC3250414 DOI: 10.1371/journal.pone.0029161] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 11/22/2011] [Indexed: 01/08/2023] Open
Abstract
Background Dermatomyositis (DM) is an autoimmune disease that mainly affects the skin, muscle, and lung. The pathogenesis of skin inflammation in DM is not well understood. Methodology and Findings We analyzed genome-wide expression data in DM skin and compared them to those from healthy controls. We observed a robust upregulation of interferon (IFN)-inducible genes in DM skin, as well as several other gene modules pertaining to inflammation, complement activation, and epidermal activation and differentiation. The interferon (IFN)-inducible genes within the DM signature were present not only in DM and lupus, but also cutaneous herpes simplex-2 infection and to a lesser degree, psoriasis. This IFN signature was absent or weakly present in atopic dermatitis, allergic contact dermatitis, acne vulgaris, systemic sclerosis, and localized scleroderma/morphea. We observed that the IFN signature in DM skin appears to be more closely related to type I than type II IFN based on in vitro IFN stimulation expression signatures. However, quantitation of IFN mRNAs in DM skin shows that the majority of known type I IFNs, as well as IFN g, are overexpressed in DM skin. In addition, both IFN-beta and IFN-gamma (but not other type I IFN) transcript levels were highly correlated with the degree of the in vivo IFN transcriptional response in DM skin. Conclusions and Significance As in the blood and muscle, DM skin is characterized by an overwhelming presence of an IFN signature, although it is difficult to conclusively define this response as type I or type II. Understanding the significance of the IFN signature in this wide array of inflammatory diseases will be furthered by identification of the nature of the cells that both produce and respond to IFN, as well as which IFN subtype is biologically active in each diseased tissue.
Collapse
Affiliation(s)
- David Wong
- Department of Dermatology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Bory Kea
- Department of Dermatology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Rob Pesich
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California, United States of America
| | - Brandon W. Higgs
- MedImmune, Translational Sciences, One MedImmune Way, Gaithersburg, Maryland, United States of America
| | - Wei Zhu
- MedImmune, Translational Sciences, One MedImmune Way, Gaithersburg, Maryland, United States of America
| | - Patrick Brown
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California, United States of America
| | - Yihong Yao
- MedImmune, Translational Sciences, One MedImmune Way, Gaithersburg, Maryland, United States of America
| | - David Fiorentino
- Department of Dermatology, Stanford University School of Medicine, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
46
|
Abstract
Alopecia areata (AA) is a nonscarring, inflammatory skin disease that results in patchy hair loss. AA is unpredictable in its onset, severity, and duration making it potentially very stressful for affected individuals. Currently, the treatment options for AA are limited and the efficacy of these treatments varies from patient to patient. The exact etiology of AA is unknown. This article provides some insights into the etiopathogenesis of AA and why some people develop it. The current knowledge on the pathogenesis of AA is summarized and some of the recent hypotheses and studies on AA are presented to allow for a fuller understanding of the possible biological mechanisms of AA.
Collapse
Affiliation(s)
- Eddy Wang
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, Canada
| | | |
Collapse
|
47
|
Haller O, Kochs G. Human MxA protein: an interferon-induced dynamin-like GTPase with broad antiviral activity. J Interferon Cytokine Res 2010; 31:79-87. [PMID: 21166595 DOI: 10.1089/jir.2010.0076] [Citation(s) in RCA: 276] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The human myxovirus resistance protein 1 (MxA) is a key mediator of the interferon-induced antiviral response against a wide range of viruses. MxA expression is tightly regulated by type I and type III interferons, requires signal transducer and activator of transcription 1 signaling, and is not inducible directly by viruses or other stimuli. MxA shares many properties with the dynamin superfamily of large GTPases. It consists of 3 domains, namely, an N-terminal GTPase domain that binds and hydrolyses GTP, a middle domain mediating self-assembly, and a carboxy-terminal GTPase effector domain. Like dynamin, MxA has the ability to self-assemble into highly ordered oligomers and to form ring-like structures around liposomes, inducing liposome tubulation. The structural details of MxA oligomerization have recently been elucidated, providing new insights into the antiviral mechanism of this mechanochemical enzyme. The structural and functional data suggest that MxA targets the nucleoprotein of MxA-sensitive viruses. Thus, MxA may form oligomeric rings around tubular nucleocapsid structures, thereby inhibiting their transcriptional and replicative function. Here we briefly review the most salient features of MxA expression and antiviral function.
Collapse
Affiliation(s)
- Otto Haller
- Department of Virology, Institute of Medical Microbiology and Hygiene, University of Freiburg, Freiburg, Germany.
| | | |
Collapse
|