1
|
IJspeert H, Edwards ESJ, O'Hehir RE, Dalm VASH, van Zelm MC. Update on inborn errors of immunity. J Allergy Clin Immunol 2024:S0091-6749(24)02407-2. [PMID: 39724969 DOI: 10.1016/j.jaci.2024.12.1075] [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: 11/15/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024]
Abstract
Ever since the first description of an inherited immunodeficiency in 1952 in a boy with gammaglobulin deficiency, new insights have progressed rapidly in disorders that are now referred to as inborn errors of immunity. In a field where fundamental molecular biology, genetics, immune signaling, and clinical care are tightly intertwined, 2022-24 saw a multitude of advances. Here we report a selection of research updates with a main focus on (1) diagnosis and screening, (2) new genetic defects, (3) susceptibility to severe coronavirus disease 2019 infection and impact of vaccination, and (4) treatment. Importantly, new pathogenic insights more rapidly affect treatment outcomes, either through an earlier and more precise diagnosis or through implementation of novel, personalized treatment. The field is growing rapidly, so awareness, communication, and collaboration are key to improving treatment outcomes.
Collapse
Affiliation(s)
- Hanna IJspeert
- Department of Clinical Genetics, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Emily S J Edwards
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Australia; Allergy, Asthma and Clinical Immunology, Alfred Health, Melbourne, Australia; Jeffrey Modell Center, Melbourne, Australia
| | - Robyn E O'Hehir
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Australia; Allergy, Asthma and Clinical Immunology, Alfred Health, Melbourne, Australia; Jeffrey Modell Center, Melbourne, Australia
| | - Virgil A S H Dalm
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands; Department of Internal Medicine, Division of Allergy & Clinical Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Menno C van Zelm
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands; Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Australia; Allergy, Asthma and Clinical Immunology, Alfred Health, Melbourne, Australia; Jeffrey Modell Center, Melbourne, Australia.
| |
Collapse
|
2
|
Tolomeo M, Cascio A. STAT4 and STAT6, their role in cellular and humoral immunity and in diverse human diseases. Int Rev Immunol 2024; 43:394-418. [PMID: 39188021 DOI: 10.1080/08830185.2024.2395274] [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: 06/29/2023] [Revised: 10/23/2023] [Accepted: 08/17/2024] [Indexed: 08/28/2024]
Abstract
Signal transducer and activator of transcription (STAT) 4 and STAT6 play a crucial role in immune cells by transducing signals from specific cytokine receptors, and inducing transcription of genes involved in cell-mediated and humoral immunity. These two different defense mechanisms against pathogens are regulated by two specific CD4+ T helper (Th) cells known as Th1 and Th2 cells. Many studies have shown that several diseases including cancer, inflammatory, autoimmune and allergic diseases are associated with a Th1/Th2 imbalance caused by increased or decreased expression/activity of STAT4 or STAT6 often due to genetic and epigenetic aberrances. An altered expression of STAT4 has been observed in different tumors and autoimmune diseases, while a dysregulation of STAT6 signaling pathway is frequently observed in allergic conditions, such as atopic dermatitis, allergic asthma, food allergy, and tumors such as Hodgkin and non-Hodgkin lymphomas. Recently, dysregulations of STAT4 and STAT6 expression have been observed in SARS-CoV2 and monkeypox infections, which are still public health emergencies in many countries. SARS-CoV-2 can induce an imbalance in Th1 and Th2 responses with a predominant activation of STAT6 in the cytosol and nuclei of pneumocytes that drives Th2 polarization and cytokine storm. In monkeypox infection the virus can promote an immune evasion by inducing a Th2 response that in turn inhibits the Th1 response essential for virus elimination. Furthermore, genetic variations of STAT4 that are associated with an increased risk of developing systemic lupus erythematosus seem to play a role in defense against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Manlio Tolomeo
- Department of Infectious Diseases, A.O.U.P. Palermo, Palermo, Italy
| | - Antonio Cascio
- Department of Infectious Diseases, A.O.U.P. Palermo, Palermo, Italy
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties, Palermo, Italy
| |
Collapse
|
3
|
Ghelich P, Samandari M, Hassani Najafabadi A, Tanguay A, Quint J, Menon N, Ghanbariamin D, Saeedinejad F, Alipanah F, Chidambaram R, Krawetz R, Nuutila K, Toro S, Barnum L, Jay GD, Schmidt TA, Tamayol A. Dissolvable Immunomodulatory Microneedles for Treatment of Skin Wounds. Adv Healthc Mater 2024; 13:e2302836. [PMID: 38299437 DOI: 10.1002/adhm.202302836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/21/2023] [Indexed: 02/02/2024]
Abstract
Sustained inflammation can halt or delay wound healing, and macrophages play a central role in wound healing. Inflammatory macrophages are responsible for the removal of pathogens, debris, and neutrophils, while anti-inflammatory macrophages stimulate various regenerative processes. Recombinant human Proteoglycan 4 (rhPRG4) is shown to modulate macrophage polarization and to prevent fibrosis and scarring in ear wound healing. Here, dissolvable microneedle arrays (MNAs) carrying rhPRG4 are engineered for the treatment of skin wounds. The in vitro experiments suggest that rhPRG4 modulates the inflammatory function of bone marrow-derived macrophages. Degradable and detachable microneedles are developed from gelatin methacryloyl (GelMA) attach to a dissolvable gelatin backing. The developed MNAs are able to deliver a high dose of rhPRG4 through the dissolution of the gelatin backing post-injury, while the GelMA microneedles sustain rhPRG4 bioavailability over the course of treatment. In vivo results in a murine model of full-thickness wounds with impaired healing confirm a decrease in inflammatory biomarkers such as TNF-α and IL-6, and an increase in angiogenesis and collagen deposition. Collectively, these results demonstrate rhPRG4-incorporating MNA is a promising platform in skin wound healing applications.
Collapse
Affiliation(s)
- Pejman Ghelich
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Mohamadmahdi Samandari
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Alireza Hassani Najafabadi
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Adam Tanguay
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Jacob Quint
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Nikhil Menon
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Delaram Ghanbariamin
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Farnoosh Saeedinejad
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Fatemeh Alipanah
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Ramaswamy Chidambaram
- Center for Comparative Medicine, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Roman Krawetz
- McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, AB, T2N 4Z6, Canada
- Department of Surgery, University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Kristo Nuutila
- US Army Institute of Surgical Research, Fort Sam Houston, Texas, 78234, USA
| | - Steven Toro
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Lindsay Barnum
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Gregory D Jay
- Emergency Medicine, Brown University, Providence, RI, 02908, USA
| | - Tannin A Schmidt
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| |
Collapse
|
4
|
Sharma M, Suratannon N, Leung D, Baris S, Takeuchi I, Samra S, Yanagi K, Rosa Duque JS, Benamar M, Del Bel KL, Momenilandi M, Béziat V, Casanova JL, van Hagen PM, Arai K, Nomura I, Kaname T, Chatchatee P, Morita H, Chatila TA, Lau YL, Turvey SE. Human germline gain-of-function in STAT6: from severe allergic disease to lymphoma and beyond. Trends Immunol 2024; 45:138-153. [PMID: 38238227 DOI: 10.1016/j.it.2023.12.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 02/12/2024]
Abstract
Signal transducer and activator of transcription (STAT)-6 is a transcription factor central to pro-allergic immune responses, although the function of human STAT6 at the whole-organism level has long remained unknown. Germline heterozygous gain-of-function (GOF) rare variants in STAT6 have been recently recognized to cause a broad and severe clinical phenotype of early-onset, multi-system allergic disease. Here, we provide an overview of the clinical presentation of STAT6-GOF disease, discussing how dysregulation of the STAT6 pathway causes severe allergic disease, and identifying possible targeted treatment approaches. Finally, we explore the mechanistic overlap between STAT6-GOF disease and other monogenic atopic disorders, and how this group of inborn errors of immunity (IEIs) powerfully inform our fundamental understanding of common human allergic disease.
Collapse
|
5
|
Catunda RQ, Ho KKY, Patel S, Roy CB, Alexiou M, Levin L, Ulrich BJ, Kaplan MH, Febbraio M. Loricrin and Cytokeratin Disorganisation in Severe Forms of Periodontitis. Int Dent J 2023; 73:862-872. [PMID: 37316411 PMCID: PMC10658443 DOI: 10.1016/j.identj.2023.05.004] [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: 12/30/2022] [Revised: 05/05/2023] [Accepted: 05/22/2023] [Indexed: 06/16/2023] Open
Abstract
OBJECTIVE The aim of this research was to investigate the role of the cornified epithelium, the outermost layer of the oral mucosa, engineered to prevent water loss and microorganism invasion, in severe forms of periodontitis (stage III or IV, grade C). METHODS Porphyromonas gingivalis, a major periodontal disease pathogen, can affect cornified epithelial protein expression through chronic activation of signal transducer and activator of transcription 6 (Stat6). We used a mouse model, Stat6VT, that mimics this to determine the effects of barrier defect on P gingivalis-induced inflammation, bone loss, and cornified epithelial protein expression, and compared histologic and immunohistologic findings with tissues obtained from human controls and patients with stage III and IV, grade C disease. Alveolar bone loss in mice was assessed using micro-computerised tomography, and soft tissue morphology was qualitatively and semi-quantitatively assessed by histologic examination for several proteins, including loricrin, filaggrin, cytokeratin 1, cytokeratin 14, a proliferation marker, a pan-leukocyte marker, as well as morphologic signs of inflammation. Relative cytokine levels were measured in mouse plasma by cytokine array. RESULTS In the tissues from patients with periodontal disease, there were greater signs of inflammation (rete pegs, clear cells, inflammatory infiltrates) and a decrease and broadening of expression of loricrin and cytokeratin 1. Cytokeratin 14 expression was also broader and decreased in stage IV. P gingivalis-infected Stat6VT mice showed greater alveolar bone loss in 9 out of 16 examined sites, and similar patterns of disruption to human patients in expression of loricrin and cytokeratins 1 and 14. There were also increased numbers of leukocytes, decreased proliferation, and greater signs of inflammation compared with P gingivalis-infected control mice. CONCLUSIONS Our study provides evidence that changes in epithelial organisation can exacerbate the effects of P gingivalis infection, with similarities to the most severe forms of human periodontitis.
Collapse
Affiliation(s)
- Raisa Queiroz Catunda
- Department of Dentistry, College of Health Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Karen Ka-Yan Ho
- Department of Dentistry, College of Health Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Srushti Patel
- Department of Dentistry, College of Health Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Christopher Bryant Roy
- Department of Dentistry, College of Health Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Maria Alexiou
- Department of Dentistry, College of Health Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Liran Levin
- Department of Dentistry, College of Health Sciences, University of Alberta, Edmonton, Alberta, Canada
| | | | - Mark H Kaplan
- Department of Microbiology & Immunology, School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Maria Febbraio
- Department of Dentistry, College of Health Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
6
|
Guttman-Yassky E, Irvine AD, Brunner PM, Kim BS, Boguniewicz M, Parmentier J, Platt AM, Kabashima K. The role of Janus kinase signaling in the pathology of atopic dermatitis. J Allergy Clin Immunol 2023; 152:1394-1404. [PMID: 37536511 DOI: 10.1016/j.jaci.2023.07.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 06/06/2023] [Accepted: 07/20/2023] [Indexed: 08/05/2023]
Abstract
Atopic dermatitis (AD) is a heterogeneous, chronic, relapsing, inflammatory skin disease associated with considerable physical, psychological, and economic burden. The pathology of AD includes complex interactions involving abnormalities in immune and skin barrier genes, skin barrier disruption, immune dysregulation, microbiome disturbance, and other environmental factors. Many of the cytokines involved in AD pathology, including IL-4, IL-13, IL-22, IL-31, thymic stromal lymphopoietin, and IFN-γ, signal through the Janus kinase (JAK)-signal transducer and activation of transcription (STAT) pathway. The JAK family includes JAK1, JAK2, JAK3, and tyrosine kinase 2; the STAT family includes STAT1, STAT2, STAT3, STAT4, STAT5A/B, and STAT6. Activation of the JAK-STAT pathway has been implicated in the pathology of several immune-mediated inflammatory diseases, including AD. However, the exact mechanisms of JAK-STAT involvement in AD have not been fully characterized. This review aims to discuss current knowledge about the role of the JAK-STAT signaling pathway and, specifically, the role of JAK1 in the pathology and symptomology of AD.
Collapse
Affiliation(s)
- Emma Guttman-Yassky
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York.
| | | | - Patrick M Brunner
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York
| | - Brian S Kim
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York
| | - Mark Boguniewicz
- Department of Pediatrics, National Jewish Health and University of Colorado School of Medicine, Denver
| | | | | | - Kenji Kabashima
- Department of Dermatology, Graduate School of Medicine, Kyoto University, Kyoto
| |
Collapse
|
7
|
Baris S, Benamar M, Chen Q, Catak MC, Martínez-Blanco M, Wang M, Fong J, Massaad MJ, Sefer AP, Kara A, Babayeva R, Eltan SB, Yucelten AD, Bozkurtlar E, Cinel L, Karakoc-Aydiner E, Zheng Y, Wu H, Ozen A, Schmitz-Abe K, Chatila TA. Severe allergic dysregulation due to a gain of function mutation in the transcription factor STAT6. J Allergy Clin Immunol 2023; 152:182-194.e7. [PMID: 36758835 PMCID: PMC10330134 DOI: 10.1016/j.jaci.2023.01.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/20/2023] [Accepted: 01/26/2023] [Indexed: 02/10/2023]
Abstract
BACKGROUND Inborn errors of immunity have been implicated in causing immune dysregulation, including allergic diseases. STAT6 is a key regulator of allergic responses. OBJECTIVES This study sought to characterize a novel gain-of-function STAT6 mutation identified in a child with severe allergic manifestations. METHODS Whole-exome and targeted gene sequencing, lymphocyte characterization, and molecular and functional analyses of mutated STAT6 were performed. RESULTS This study reports a child with a missense mutation in the DNA binding domain of STAT6 (c.1114G>A, p.E372K) who presented with severe atopic dermatitis, eosinophilia, and elevated IgE. Naive lymphocytes from the affected patient displayed increased TH2- and suppressed TH1- and TH17-cell responses. The mutation augmented both basal and cytokine-induced STAT6 phosphorylation without affecting dephosphorylation kinetics. Treatment with the Janus kinase 1/2 inhibitor ruxolitinib reversed STAT6 hyperresponsiveness to IL-4, normalized TH1 and TH17 cells, suppressed the eosinophilia, and improved the patient's atopic dermatitis. CONCLUSIONS This study identified a novel inborn error of immunity due to a STAT6 gain-of-function mutation that gave rise to severe allergic dysregulation. Janus kinase inhibitor therapy could represent an effective targeted treatment for this disorder.
Collapse
Affiliation(s)
- Safa Baris
- Division of Pediatric Allergy and Immunology School of Medicine, Marmara University, Istanbul, Turkey; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey; The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Mehdi Benamar
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Qian Chen
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Mehmet Cihangir Catak
- Division of Pediatric Allergy and Immunology School of Medicine, Marmara University, Istanbul, Turkey; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey; The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Mónica Martínez-Blanco
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Muyun Wang
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Jason Fong
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Michel J Massaad
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut, Lebanon; Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Asena Pinar Sefer
- Division of Pediatric Allergy and Immunology School of Medicine, Marmara University, Istanbul, Turkey; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey; The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Altan Kara
- TUBITAK Marmara Research Center, Gene Engineering and Biotechnology Institute, Gebze, Turkey
| | - Royala Babayeva
- Division of Pediatric Allergy and Immunology School of Medicine, Marmara University, Istanbul, Turkey; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey; The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Sevgi Bilgic Eltan
- Division of Pediatric Allergy and Immunology School of Medicine, Marmara University, Istanbul, Turkey; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey; The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Ayse Deniz Yucelten
- Department of Dermatology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Emine Bozkurtlar
- Department of Pathology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Leyla Cinel
- Department of Pathology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Elif Karakoc-Aydiner
- Division of Pediatric Allergy and Immunology School of Medicine, Marmara University, Istanbul, Turkey; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey; The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Yumei Zheng
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Mass; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Mass
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Mass; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Mass
| | - Ahmet Ozen
- Division of Pediatric Allergy and Immunology School of Medicine, Marmara University, Istanbul, Turkey; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey; The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Klaus Schmitz-Abe
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass; The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Mass
| | - Talal A Chatila
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass.
| |
Collapse
|
8
|
Comparison of cytokine mediators in type 2 inflammatory conditions on the skin and ocular surface. Curr Opin Allergy Clin Immunol 2022; 22:319-327. [DOI: 10.1097/aci.0000000000000842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
9
|
Khan IM, Ulrich BJ, Nelson AS, Sehra S, Kansas GS, Kaplan MH. Selectin Dependence of Allergic Skin Inflammation Is Diminished by Maternal Atopy. Immunohorizons 2021; 5:703-710. [PMID: 34433625 PMCID: PMC8638165 DOI: 10.4049/immunohorizons.2100052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 08/04/2021] [Indexed: 11/19/2022] Open
Abstract
Allergic skin inflammation requires the influx of inflammatory cells into the skin. Extravasation of leukocytes into the skin requires interactions between endothelial selectins and their glycan ligands on the surface of leukocytes. Selectin-ligand formation requires the activity of several glycosyltransferases, including Fut7 In this report, we tested the importance of Fut7 for the development of allergic skin inflammation in the Stat6VT transgenic mouse model. We observed that Fut7 deficiency was protective but did not eliminate disease. Segregation of the data by gender of the parent that transmitted the Stat6VT transgene, but not by gender of the pups, which were analyzed for disease, revealed that the protective effects of Fut7 deficiency were significantly greater when dams were Stat6VT negative. In contrast, in mice from litters of Stat6VT+ dams, Fut7 deficiency resulted in only modest protection. These findings indicate that pups from atopic dams exhibit a greater propensity for allergic disease, similar to observations in humans, and that the effect of maternal atopy is due to enhanced selectin-independent mechanisms of leukocyte recruitment in their offspring. Together, these results demonstrate that Fut7 deficiency can be protective in a model of atopic dermatitis but that maternal atopy diminishes these protective effects, suggesting alternative pathways for leukocyte recruitment in the absence of Fut7 enzyme activity. These observations have implications for understanding how the environment in utero predisposes for the development of allergic disease.
Collapse
Affiliation(s)
- Ibrahim M Khan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
| | - Benjamin J Ulrich
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
| | - Andrew S Nelson
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN; and
| | - Sarita Sehra
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN; and
| | - Geoffrey S Kansas
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Mark H Kaplan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN;
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN; and
| |
Collapse
|
10
|
Translational Relevance of Mouse Models of Atopic Dermatitis. J Clin Med 2021; 10:jcm10040613. [PMID: 33561938 PMCID: PMC7914954 DOI: 10.3390/jcm10040613] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/31/2021] [Accepted: 02/03/2021] [Indexed: 02/06/2023] Open
Abstract
The complexity of atopic dermatitis (AD) continues to present a challenge in the appropriate selection of a mouse model because no single murine model completely recapitulates all aspects of human AD. This has been further complicated by recent evidence of the distinct AD endotypes that are dictated by unique patterns of inflammation involving Th1, Th2, Th17, and Th22 axes. A review of currently used mouse models demonstrates that while all AD mouse models consistently exhibit Th2 inflammation, only some demonstrate concomitant Th17 and/or Th22 induction. As the current understanding of the pathogenic contributions of these unique endotypes and their potential therapeutic roles expands, ongoing efforts to maximize a given mouse model’s homology with human AD necessitates a close evaluation of its distinct immunological signature.
Collapse
|
11
|
Lee E, Ahn S, Jin SH, Lee M, Pyo JJ, Shin JC, An S, Ha J, Noh M. CXCL14 downregulation in human keratinocytes is a potential biomarker for a novel in vitro skin sensitization test. Toxicol Appl Pharmacol 2020; 386:114828. [DOI: 10.1016/j.taap.2019.114828] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 11/13/2019] [Accepted: 11/13/2019] [Indexed: 01/21/2023]
|
12
|
Interleukin-Mediated Pendrin Transcriptional Regulation in Airway and Esophageal Epithelia. Int J Mol Sci 2019; 20:ijms20030731. [PMID: 30744098 PMCID: PMC6386862 DOI: 10.3390/ijms20030731] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/29/2019] [Accepted: 01/29/2019] [Indexed: 12/28/2022] Open
Abstract
Pendrin (SLC26A4), a Cl−/anion exchanger, is expressed at high levels in kidney, thyroid, and inner ear epithelia, where it has an essential role in bicarbonate secretion/chloride reabsorption, iodide accumulation, and endolymph ion balance, respectively. Pendrin is expressed at lower levels in other tissues, such as airways and esophageal epithelia, where it is transcriptionally regulated by the inflammatory cytokines interleukin (IL)-4 and IL-13 through a signal transducer and activator of transcription 6 (STAT6)-mediated pathway. In the airway epithelium, increased pendrin expression during inflammatory diseases leads to imbalances in airway surface liquid thickness and mucin release, while, in the esophageal epithelium, dysregulated pendrin expression is supposed to impact the intracellular pH regulation system. In this review, we discuss some of the recent findings on interleukin-mediated transcriptional regulation of pendrin and how this dysregulation impacts airway and esophagus epithelial homeostasis during inflammatory diseases.
Collapse
|
13
|
Benmoussa K, Garaude J, Acín-Pérez R. How Mitochondrial Metabolism Contributes to Macrophage Phenotype and Functions. J Mol Biol 2018; 430:3906-3921. [PMID: 30006265 DOI: 10.1016/j.jmb.2018.07.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 07/02/2018] [Accepted: 07/03/2018] [Indexed: 01/02/2023]
Abstract
Metabolic reprogramming of cells from the innate immune system is one of the most noteworthy topics in immunological research nowadays. Upon infection or tissue damage, innate immune cells, such as macrophages, mobilize various immune and metabolic signals to mount a response best suited to eradicate the threat. Current data indicate that both the immune and metabolic responses are closely interconnected. On account of its peculiar position in regulating both of these processes, the mitochondrion has emerged as a critical organelle that orchestrates the coordinated metabolic and immune adaptations in macrophages. Significant effort is now underway to understand how metabolic features of differentiated macrophages regulate their immune specificities with the eventual goal to manipulate cellular metabolism to control immunity. In this review, we highlight some of the recent work that place cellular and mitochondrial metabolism in a central position in the macrophage differentiation program.
Collapse
Affiliation(s)
- Khaddouj Benmoussa
- Laboratoire Maladies Rares, Génétique et Métabolisme, INSERM U1211, Université de Bordeaux, CHU Pellegrin, École de Sages-Femmes, 33000 Bordeaux, France
| | - Johan Garaude
- Laboratoire Maladies Rares, Génétique et Métabolisme, INSERM U1211, Université de Bordeaux, CHU Pellegrin, École de Sages-Femmes, 33000 Bordeaux, France.
| | - Rebeca Acín-Pérez
- UCLA Division of Endocrinology, Diabetes and Metabolism, David Geffen School of Medicine, 10833 Le Conte Avenue, CHS 27-200, Los Angeles, CA 90025, USA; Centro Nacional de Investigaciones Cardiovasculares Carlos III, Melchor Fernandez de Almagro 3, 28029 Madrid, Spain.
| |
Collapse
|
14
|
DaSilva-Arnold SC, Thyagarajan A, Seymour LJ, Yi Q, Bradish JR, Al-Hassani M, Zhou H, Perdue NJ, Nemeth V, Krbanjevic A, Serezani APM, Olson MR, Spandau DF, Travers JB, Kaplan MH, Turner MJ. Phenotyping acute and chronic atopic dermatitis-like lesions in Stat6VT mice identifies a role for IL-33 in disease pathogenesis. Arch Dermatol Res 2018; 310:197-207. [PMID: 29368135 PMCID: PMC6198812 DOI: 10.1007/s00403-018-1807-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 12/18/2017] [Accepted: 01/04/2018] [Indexed: 12/27/2022]
Abstract
The Stat6VT mouse model of atopic dermatitis (AD) is induced by T-cell-specific expression of a constitutively active form of the protein signal transducer and activator of transcription 6 (STAT6). Although AD-like lesions are known to develop in Stat6VT mice, this study was designed to determine if these mice develop acute and chronic phases of disease similar to humans. To address this, AD-like lesions from Stat6VT mice were harvested at two different timepoints relative to their onset. Lesions harvested within 1 week after development were defined as acute lesions, and those present for 1 month or more were defined as chronic lesions. Acute and chronic AD-like lesions from Stat6VT mice exhibited histologic findings and cytokine expression patterns similar to acute and chronic AD lesions in humans. Further analysis revealed increased levels of interleukin (IL)-33 transcripts in AD-like lesions compared to Stat6VT nonlesional and wild-type skin controls. Immunofluorescence also revealed increased numbers of IL-33+ keratinocytes in Stat6VT lesional skin and localized IL-33+ keratinocytes to a keratin 5+ subset. Furthermore, AD-like disease was more severe in IL-33-deficient Stat6VT mice compared to IL-33-sufficient Stat6VT mice. These studies suggest that Stat6VT mice can serve as a model of acute and chronic AD and that IL-33 may attenuate inflammation in this system.
Collapse
Affiliation(s)
- Sonia C DaSilva-Arnold
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Anita Thyagarajan
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Leroy J Seymour
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Qiaofang Yi
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Joshua R Bradish
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Mohammed Al-Hassani
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Hongming Zhou
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Nikolajs J Perdue
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Val Nemeth
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Aleksandar Krbanjevic
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Ana P M Serezani
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Matthew R Olson
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Dan F Spandau
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jeffrey B Travers
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, USA
| | - Mark H Kaplan
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Matthew J Turner
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, USA.
| |
Collapse
|
15
|
Zaffini R, Gotte G, Menegazzi M. Asthma and poly(ADP-ribose) polymerase inhibition: a new therapeutic approach. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:281-293. [PMID: 29483769 PMCID: PMC5813949 DOI: 10.2147/dddt.s150846] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Asthma is a chronic lung disease affecting people of all ages worldwide, and it frequently begins in childhood. Because of its chronic nature, it is characterized by pathological manifestations, including airway inflammation, remodeling, and goblet cell hyperplasia. Current therapies for asthma, including corticosteroids and beta-2 adrenergic agonists, are directed toward relieving the symptoms of the asthmatic response, with poor effectiveness against the underlying causes of the disease. Asthma initiation and progression depends on the T helper (Th) 2 type immune response carried out by a complex interplay of cytokines, such as interleukin (IL) 4, IL5, and IL13, and the signal transducer and activator of transcription 6. Much of the data resulting from different laboratories support the role of poly(ADP-ribose) polymerase (PARP) 1 and PARP14 activation in asthma. Indeed, PARP enzymes play key roles in the regulation and progression of the inflammatory asthma process because they affect the expression of genes and chemokines involved in the immune response. Consistently, PARP inhibition achievable either upon genetic ablation or by using pharmacological agents has shown a range of therapeutic effects against the disease. Indeed, in the last two decades, several preclinical studies highlighted the protective effects of PARP inhibition in various animal models of asthma. PARP inhibitors showed the ability to reduce the overall lung inflammation acting with a specific effect on immune cell recruitment and through the modulation of asthma-associated cytokines production. PARP inhibition has been shown to affect the Th1–Th2 balance and, at least in some aspects, the airway remodeling. In this review, we summarize and discuss the steps that led PARP inhibition to become a possible future therapeutic strategy against allergic asthma.
Collapse
Affiliation(s)
- Raffaela Zaffini
- Department of Neuroscience, Biomedicine and Movement Science, Biochemistry Section, University of Verona, Verona, Italy
| | - Giovanni Gotte
- Department of Neuroscience, Biomedicine and Movement Science, Biochemistry Section, University of Verona, Verona, Italy
| | - Marta Menegazzi
- Department of Neuroscience, Biomedicine and Movement Science, Biochemistry Section, University of Verona, Verona, Italy
| |
Collapse
|
16
|
IL-4 driven transcription factor FoxQ1 is expressed by monocytes in atopic dermatitis and stimulates monocyte migration. Sci Rep 2017; 7:16847. [PMID: 29203829 PMCID: PMC5715145 DOI: 10.1038/s41598-017-17307-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 11/22/2017] [Indexed: 12/18/2022] Open
Abstract
Monocytes are actively recruited at sites of chronic inflammation. However, molecular factors involved in this process are not fully elucidated. Here, we show that cytokine IL-4 which is implicated in the development of chronic inflammatory disease atopic dermatitis (AD) induces expression of transcription factor FoxQ1 in human monocytes and macrophages. FoxQ1 mRNA levels were elevated in monocytes of AD patients compared to healthy donors. Overexpression of FoxQ1 in RAW 264.7 monocytic cells facilitated their migration towards MCP-1 and was associated with decreased expression of migration-regulating genes (claudin 11 and plexin C1). Furthermore, FoxQ1 overexpression in RAW cells accelerated TNFα secretion after LPS challenge. Overall, our results indicate that FoxQ1 stimulates monocyte motility, increases pro-inflammatory potential, and directs monocyte migration towards MCP-1 that is crucial for monocyte influx into inflammatory sites. This mechanism could contribute to the pathogenesis of chronic inflammatory disorders such as AD.
Collapse
|
17
|
Kälin S, Becker M, Ott VB, Serr I, Hosp F, Mollah MMH, Keipert S, Lamp D, Rohner-Jeanrenaud F, Flynn VK, Scherm MG, Nascimento LFR, Gerlach K, Popp V, Dietzen S, Bopp T, Krishnamurthy P, Kaplan MH, Serrano M, Woods SC, Tripal P, Palmisano R, Jastroch M, Blüher M, Wolfrum C, Weigmann B, Ziegler AG, Mann M, Tschöp MH, Daniel C. A Stat6/Pten Axis Links Regulatory T Cells with Adipose Tissue Function. Cell Metab 2017; 26:475-492.e7. [PMID: 28877454 PMCID: PMC5627977 DOI: 10.1016/j.cmet.2017.08.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 07/21/2017] [Accepted: 08/08/2017] [Indexed: 12/22/2022]
Abstract
Obesity and type 2 diabetes are associated with metabolic defects and adipose tissue inflammation. Foxp3+ regulatory T cells (Tregs) control tissue homeostasis by counteracting local inflammation. However, if and how T cells interlink environmental influences with adipocyte function remains unknown. Here, we report that enhancing sympathetic tone by cold exposure, beta3-adrenergic receptor (ADRB3) stimulation or a short-term high-calorie diet enhances Treg induction in vitro and in vivo. CD4+ T cell proteomes revealed higher expression of Foxp3 regulatory networks in response to cold or ADRB3 stimulation in vivo reflecting Treg induction. Specifically, Ragulator-interacting protein C17orf59, which limits mTORC1 activity, was upregulated in CD4+ T cells by either ADRB3 stimulation or cold exposure, suggesting contribution to Treg induction. By loss- and gain-of-function studies, including Treg depletion and transfers in vivo, we demonstrated that a T cell-specific Stat6/Pten axis links cold exposure or ADRB3 stimulation with Foxp3+ Treg induction and adipose tissue function. Our findings offer a new mechanistic model in which tissue-specific Tregs maintain adipose tissue function.
Collapse
Affiliation(s)
- Stefanie Kälin
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München and Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 85748 Munich, Germany; German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany
| | - Maike Becker
- German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; Institute for Diabetes Research, Research Group Immune Tolerance in Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany
| | - Verena B Ott
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München and Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 85748 Munich, Germany; German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; Institute for Advanced Study, Technische Universität München, 85748 Garching, Germany
| | - Isabelle Serr
- German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; Institute for Diabetes Research, Research Group Immune Tolerance in Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany
| | - Fabian Hosp
- Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Mohammad M H Mollah
- German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; Institute for Diabetes Research, Research Group Immune Tolerance in Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany
| | - Susanne Keipert
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München and Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 85748 Munich, Germany; German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany
| | - Daniel Lamp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München and Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 85748 Munich, Germany; German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany
| | - Francoise Rohner-Jeanrenaud
- Laboratory of Metabolism, Division of Endocrinology, Diabetology, Hypertension, and Nutrition, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Victoria K Flynn
- German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; Institute for Diabetes Research, Research Group Immune Tolerance in Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany
| | - Martin G Scherm
- German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; Institute for Diabetes Research, Research Group Immune Tolerance in Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany
| | - Lucas F R Nascimento
- German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; Institute for Diabetes Research, Research Group Immune Tolerance in Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany
| | - Katharina Gerlach
- Department of Medicine 1, University of Erlangen-Nuremberg, 91052 Erlangen, Germany
| | - Vanessa Popp
- Department of Medicine 1, University of Erlangen-Nuremberg, 91052 Erlangen, Germany
| | - Sarah Dietzen
- Institute of Immunology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Tobias Bopp
- Institute of Immunology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Purna Krishnamurthy
- Department of Pediatrics and HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Mark H Kaplan
- Department of Pediatrics and HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Manuel Serrano
- Tumour Suppression Group, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Stephen C Woods
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA
| | - Philipp Tripal
- OICE (Optical Imaging Centre Erlangen), University Erlangen, 91052 Erlangen, Germany
| | - Ralf Palmisano
- OICE (Optical Imaging Centre Erlangen), University Erlangen, 91052 Erlangen, Germany
| | - Martin Jastroch
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München and Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 85748 Munich, Germany; German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany
| | - Matthias Blüher
- Department of Medicine, Research Group Molecular Endocrinology, University of Leipzig, 04103 Leipzig, Germany
| | - Christian Wolfrum
- Swiss Federal Institute of Technology, Institute of Food Nutrition and Health, Laboratory of Translational Nutrition Biology, ETH Zurich, 8603 Schwerzenbach, Switzerland
| | - Benno Weigmann
- Department of Medicine 1, University of Erlangen-Nuremberg, 91052 Erlangen, Germany
| | - Anette-Gabriele Ziegler
- German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany; Institute for Diabetes Research, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany; Klinikum rechts der Isar, Technische Universität München, 80333 Munich, Germany
| | - Matthias Mann
- Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München and Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 85748 Munich, Germany; German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany.
| | - Carolin Daniel
- Institute for Diabetes Research, Research Group Immune Tolerance in Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany; German Center for Diabetes Research (DZD), 85764 Munich-Neuherberg, Germany.
| |
Collapse
|
18
|
Knol EF, Hijnen D. Atopic dermatitis: A tale of two distinct pathomechanisms that make you itch. Eur J Immunol 2017; 46:2512-2515. [PMID: 27813070 DOI: 10.1002/eji.201646708] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 09/27/2016] [Accepted: 10/07/2016] [Indexed: 02/01/2023]
Abstract
Atopic dermatitis (AD) or eczema is the most common chronic inflammatory skin disease. It is a multifactorial disease with local and systemic immune changes. Current therapies focus on restoring the local skin barrier or inhibiting immune responses. In this issue of the European Journal of Immunology, Sehra et al. [Eur. J. Immunol. 2016. 46:2609-2613] describe a mouse model with T-cell-specific expression of constitutively active Stat6 in Flaky tail mice, which have mutations in the Flg and Tmem79 genes. The authors describe that it is the combination of changes in the skin barrier proteins filaggrin and Tmem79, together with Th2 cytokine signaling in the constitutively active Stat6 transgene, that drives the immune-pathomechanism in AD. These results are consistent with human studies where it is demonstrated that diminished filaggrin expression in skin is a predisposing factor for AD, but is neither required nor sufficient for disease indicating that additional factors are required for disease development. The current mouse model by Sehra et al. could be instrumental in evaluation new therapeutic strategies for AD.
Collapse
Affiliation(s)
- Edward F Knol
- Department of Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands. .,Department of Dermatology and Allergology, University Medical Centre Utrecht, Utrecht, The Netherlands.
| | - DirkJan Hijnen
- Department of Dermatology and Allergology, University Medical Centre Utrecht, Utrecht, The Netherlands
| |
Collapse
|
19
|
Krishnamurthy P, Da-Silva-Arnold S, Turner MJ, Travers JB, Kaplan MH. Poly-ADP ribose polymerase-14 limits severity of allergic skin disease. Immunology 2017; 152:451-461. [PMID: 28653395 DOI: 10.1111/imm.12782] [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: 04/21/2017] [Revised: 06/02/2017] [Accepted: 06/21/2017] [Indexed: 12/31/2022] Open
Abstract
Poly-ADP ribose polymerase-14 (PARP14 or ARTD8) was initially identified as a transcriptional co-activator for signal transducer and activator of transcription 6 (Stat6), where the presence of interleukin-4 (IL-4) and activated Stat6 induces the enzymatic activity of PARP14 that promotes T helper type 2 differentiation and allergic airway disease. To further our understanding of PARP14 in allergic disease, we studied the function of PARP14 in allergic inflammation of skin using mice that express constitutively active Stat6 in T cells (Stat6VT) and develop spontaneous inflammation of the skin. We mated Stat6VT mice to Parp14-/- mice and observed that approximately 75% of the Stat6VT × Parp14-/- mice develop severe atopic dermatitis (AD)-like lesions, compared with about 50% of Stat6VT mice, and have increased morbidity compared with Stat6VT mice. Despite this, gene expression in the skin and the cellular infiltrates was only modestly altered by the absence of PARP14. In contrast, we saw significant changes in systemic T-cell cytokine production. Moreover, adoptive transfer experiments demonstrated that decreases in IL-4 production reflected a cell intrinsic role for PARP14 in Th2 cytokine control. Hence, our data suggest that although PARP14 has similar effects on T-cell cytokine production in several allergic disease models, the outcome of those effects is distinct, depending on the target organ of disease.
Collapse
Affiliation(s)
- Purna Krishnamurthy
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sonia Da-Silva-Arnold
- Department of Dermatology, and Roudebush Veterans' Administration Hospital, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Matthew J Turner
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Dermatology, and Roudebush Veterans' Administration Hospital, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jeffrey B Travers
- Department of Dermatology, and Roudebush Veterans' Administration Hospital, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Pharmacology and Toxicology, Wright State University, Dayton, OH, USA
| | - Mark H Kaplan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Pediatrics and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
20
|
Goswami R, Kaplan M. STAT Transcription Factors in T Cell Control of Health and Disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 331:123-180. [DOI: 10.1016/bs.ircmb.2016.09.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
21
|
Di Mise A, Wang YX, Zheng YM. Role of Transcription Factors in Pulmonary Artery Smooth Muscle Cells: An Important Link to Hypoxic Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 967:13-32. [PMID: 29047078 DOI: 10.1007/978-3-319-63245-2_2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hypoxia, namely a lack of oxygen in the blood, induces pulmonary vasoconstriction and vasoremodeling, which serve as essential pathologic factors leading to pulmonary hypertension (PH). The underlying molecular mechanisms are uncertain; however, pulmonary artery smooth muscle cells (PASMCs) play an essential role in hypoxia-induced pulmonary vasoconstriction, vasoremodeling, and PH. Hypoxia causes oxidative damage to DNAs, proteins, and lipids. This damage (oxidative stress) modulates the activity of ion channels and elevates the intracellular calcium concentration ([Ca2+]i, Ca2+ signaling) of PASMCs. The oxidative stress and increased Ca2+ signaling mutually interact with each other, and synergistically results in a variety of cellular responses. These responses include functional and structural abnormalities of mitochondria, sarcoplasmic reticulum, and nucleus; cell contraction, proliferation, migration, and apoptosis, as well as generation of vasoactive substances, inflammatory molecules, and growth factors that mediate the development of PH. A number of studies reveal that various transcription factors (TFs) play important roles in hypoxia-induced oxidative stress, disrupted PAMSC Ca2+ signaling and the development and progress of PH. It is believed that in the pathogenesis of PH, hypoxia facilitates these roles by mediating the expression of multiple genes. Therefore, the identification of specific genes and their transcription factors implicated in PH is necessary for the complete understanding of the underlying molecular mechanisms. Moreover, this identification may aid in the development of novel and effective therapeutic strategies for PH.
Collapse
Affiliation(s)
- Annarita Di Mise
- Department of Molecular & Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - Yong-Xiao Wang
- Department of Molecular & Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA.
| | - Yun-Min Zheng
- Department of Molecular & Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA.
| |
Collapse
|
22
|
Sehra S, Krishnamurthy P, Koh B, Zhou HM, Seymour L, Akhtar N, Travers JB, Turner MJ, Kaplan MH. Increased Th2 activity and diminished skin barrier function cooperate in allergic skin inflammation. Eur J Immunol 2016; 46:2609-2613. [PMID: 27510401 DOI: 10.1002/eji.201646421] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 06/30/2016] [Accepted: 08/08/2016] [Indexed: 12/31/2022]
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease induced by a complex interaction between susceptibility genes encoding skin barrier components and environmental allergen exposure that results in type 2 cytokine production. Although genetic lesions in either component can be risk factors for disease in patients, whether these pathways interact in the development of AD is not clear. To test this, we mated mice with T-cell specific expression of constitutively active Stat6 (Stat6VT) that spontaneously develop allergic skin inflammation with Flaky tail (Ft) mice that have mutations in Flg and Tmem79 genes that each affect skin barrier function. Our results demonstrate that over 90% of the Stat6VT transgenic mice carrying the Ft alleles (Stat6VTxFt-/- ) develop severe atopic dermatitis lesions by 3-5 months of age, compared with only 40% of Stat6VT mice that develop disease by 6-7 months of age. Further, histopathological analysis of skin tissues from Stat6VTxFt-/- mice revealed extensive thickening of the dermis with increased inflammatory infiltrates as compared with Stat6VT mice. Our study suggests that skin barrier defects and altered Th2 responses independently cooperate in the pathogenesis of allergic skin inflammation, similar to effects observed in patients with AD.
Collapse
Affiliation(s)
- Sarita Sehra
- Department of Pediatrics and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Purna Krishnamurthy
- Department of Pediatrics and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Byunghee Koh
- Department of Pediatrics and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Hong-Ming Zhou
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lee Seymour
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nahid Akhtar
- Department of Pediatrics and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jeffrey B Travers
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Pharmacology and Toxicology, Wright State University, Dayton, OH, USA
| | - Matthew J Turner
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mark H Kaplan
- Department of Pediatrics and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA. .,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
23
|
Krishnamurthy P, Kaplan MH. STAT6 and PARP Family Members in the Development of T Cell-dependent Allergic Inflammation. Immune Netw 2016; 16:201-10. [PMID: 27574499 PMCID: PMC5002446 DOI: 10.4110/in.2016.16.4.201] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/17/2016] [Accepted: 07/20/2016] [Indexed: 12/11/2022] Open
Abstract
Allergic inflammation requires the orchestration of altered gene expression in the target tissue and in the infiltrating immune cells. The transcription factor STAT6 is critical in activating cytokine gene expression and cytokine signaling both in the immune cells and in target tissue cells including airway epithelia, keratinocytes and esophageal epithelial cells. STAT6 is activated by the cytokines IL-4 and IL-13 to mediate the pathogenesis of allergic disorders such as asthma, atopic dermatitis, food allergy and eosinophilic esophagitis (EoE). In this review, we summarize the role of STAT6 in allergic diseases, its interaction with the co-factor PARP14 and the molecular mechanisms by which STAT6 and PARP14 regulate gene transcription.
Collapse
Affiliation(s)
- Purna Krishnamurthy
- Department of Pediatrics, Wells Center for Pediatric Research, Indianapolis, IN 46202, USA.; Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Mark H Kaplan
- Department of Pediatrics, Wells Center for Pediatric Research, Indianapolis, IN 46202, USA.; Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
24
|
Kantor R, Silverberg JI. Environmental risk factors and their role in the management of atopic dermatitis. Expert Rev Clin Immunol 2016; 13:15-26. [PMID: 27417220 DOI: 10.1080/1744666x.2016.1212660] [Citation(s) in RCA: 209] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION The etiology of atopic dermatitis (AD) is multifactorial with interaction between genetics, immune and environmental factors. Areas covered: We review the role of prenatal exposures, irritants and pruritogens, pathogens, climate factors, including temperature, humidity, ultraviolet radiation, outdoor and indoor air pollutants, tobacco smoke exposure, water hardness, urban vs. rural living, diet, breastfeeding, probiotics and prebiotics on AD. Expert commentary: The increased global prevalence of AD cannot be attributed to genetics alone, suggesting that evolving environmental exposures may trigger and/or flare disease in predisposed individuals. There is a complex interplay between different environmental factors, including individual use of personal care products and exposure to climate, pollution, food and other exogenous factors. Understanding these complex risk factors is crucial to developing targeted interventions to prevent the disease in millions. Moreover, patients require counseling on optimal regimens for minimization of exposure to irritants and pruritogens and other harmful exposures.
Collapse
Affiliation(s)
- Robert Kantor
- a Department of Dermatology , Northwestern University , Chicago , IL , USA
| | - Jonathan I Silverberg
- b Departments of Dermatology, Preventive Medicine and Medical Social Sciences , Northwestern University , Chicago , IL , USA
| |
Collapse
|
25
|
Sehra S, Serezani APM, Ocaña JA, Travers JB, Kaplan MH. Mast Cells Regulate Epidermal Barrier Function and the Development of Allergic Skin Inflammation. J Invest Dermatol 2016; 136:1429-1437. [PMID: 27021404 DOI: 10.1016/j.jid.2016.03.019] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 02/15/2016] [Accepted: 03/06/2016] [Indexed: 11/30/2022]
Abstract
Atopic dermatitis is a chronic inflammatory skin disease characterized by infiltration of eosinophils, T helper cells, and mast cells. The role of mast cells in atopic dermatitis is not completely understood. To define the effects of mast cells on skin biology, we observed that mast cells regulate the homeostatic expression of epidermal differentiation complex and other skin genes. Decreased epidermal differentiation complex gene expression in mice that genetically lack mast cells (Kit(W-sh/W-sh) mice) is associated with increased uptake of protein antigens painted on the skin by dendritic cells (DCs) compared with similarly treated wild-type mice, suggesting a protective role for mast cells in exposure to nominal environmental allergens. To test this further, we crossed Kit(W-sh/W-sh) mice with signal transducer and activator of transcription 6 (i.e., Stat6) VT transgenic mice that develop spontaneous atopic dermatitis-like disease that is dependent on T helper cell 2 cytokines and is associated with high serum concentrations of IgE. We observed that Stat6VT × Kit(W-sh/W-sh) mice developed more frequent and more severe allergic skin inflammation than Stat6VT transgenic mice that had mast cells. Together, these studies suggest that mast cells regulate epidermal barrier function and have a potential protective role in the development of atopic dermatitis-like disease.
Collapse
Affiliation(s)
- Sarita Sehra
- Department of Pediatrics and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ana P M Serezani
- Department of Pediatrics and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jesus A Ocaña
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jeffrey B Travers
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA; Department of Dermatology, Indiana University School of Medicine, Indianapolis, Indiana, USA; Department of Pharmacology and Toxicology, Wright State University, Dayton, Ohio, USA
| | - Mark H Kaplan
- Department of Pediatrics and Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA; Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| |
Collapse
|
26
|
Hoppenot D, Malakauskas K, Lavinskiene S, Sakalauskas R. p-STAT6, PU.1, and NF-κB are involved in allergen-induced late-phase airway inflammation in asthma patients. BMC Pulm Med 2015; 15:122. [PMID: 26466682 PMCID: PMC4606997 DOI: 10.1186/s12890-015-0119-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 10/06/2015] [Indexed: 11/23/2022] Open
Abstract
Background Previous in vitro and animal studies demonstrated that transcription factors p-STAT6 and PU.1 are required to induce interleukin (IL)-9 secretion by T helper (Th) 9 cells. It is believed that n factor-kappaB (NF-κB) plays a role in eosinophil survival. The importance of these transcription factors in the pathogenesis of allergic asthma (AA) in humans is poorly understood. We evaluated p-STAT6 and PU.1 expression in peripheral blood Th9 cells and NF-κB expression in eosinophils during late-phase airway inflammation in AA patients. Methods Nineteen adults with AA and 14 adult healthy individuals (HI) were examined. Peripheral blood collected 24 h before (baseline) and 24 h after bronchial allergen challenge. CD4+ cells and eosinophils were isolated by high-density gradient centrifugation and magnetic separation. The percentage of Th9 cells and apoptotic eosinophils was estimated by flow cytometry. p-STAT6 and PU.1 expression was expressed as mean fluorescence intensity (MFI) in Th9 cells. NF-κB levels were expressed as MFI in peripheral blood eosinophils. Serum IL-9 and IL-5 levels were determined by enzyme-linked immunosorbent assay. Results At baseline, MFI of p-STAT6 and PU.1 in peripheral blood Th9 cells and MFI of NF-κB in eosinophils and, serum IL-5 and IL-9 levels were greater in AA patients (P < 0.05). Decreased eosinophil apoptosis was seen in the AA group compared with HI (P < 0.05). MFI of p-STAT6, PU.1, and NF-κB and serum levels of IL-5 and IL-9 were increased in the AA group 24 h after challenge compared with baseline (P < 0.05). In the AA group, a correlation between serum IL-9 and Th9 cells (r = 0.7, P = 0.001) and MFI of PU.1 (r = 0.6, P = 0.01) 24 h after bronchial allergen challenge was observed. A correlation between Th9 cells and MFI of p-STAT6 (r = 0.45, P = 0.03) as well as MFI of PU.1 (r = 0.5, P = 0.02) 24 h after challenge was only observed in AA patients. A correlation between the MFI of NF-κB and eosinophil apoptosis was observed in AA patients 24 h before (r = −0.46, P = 0.02) and after (r = −0.5, P = 0.02) challenge. Discussions p-STAT6 and PU.1 may be associated with Th9 cells and IL-9 production, whereas NF-κB and IL-5 may be associated with reduced eosinophil apoptosis in allergen-induced late-phase airway inflammation. Trial registration ClinicalTrials.gov NCT02214303 Electronic supplementary material The online version of this article (doi:10.1186/s12890-015-0119-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Deimante Hoppenot
- Department of Pulmonology and Immunology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania.
| | - Kestutis Malakauskas
- Department of Pulmonology and Immunology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania. .,Laboratory of Pulmonology, Department of Pulmonology and Immunology, Medical Academ, Lithuanian University of Health Sciences, Kaunas, Lithuania.
| | - Simona Lavinskiene
- Department of Pulmonology and Immunology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania. .,Laboratory of Pulmonology, Department of Pulmonology and Immunology, Medical Academ, Lithuanian University of Health Sciences, Kaunas, Lithuania.
| | - Raimundas Sakalauskas
- Department of Pulmonology and Immunology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania.
| |
Collapse
|
27
|
Levy LL, Urban J, King BA. Treatment of recalcitrant atopic dermatitis with the oral Janus kinase inhibitor tofacitinib citrate. J Am Acad Dermatol 2015; 73:395-9. [PMID: 26194706 DOI: 10.1016/j.jaad.2015.06.045] [Citation(s) in RCA: 153] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 06/16/2015] [Accepted: 06/24/2015] [Indexed: 12/30/2022]
Abstract
BACKGROUND Treatment of moderate to severe atopic dermatitis (AD) is often inadequate. OBJECTIVE We sought to evaluate the efficacy of the oral Janus kinase inhibitor tofacitinib citrate in the treatment of moderate to severe AD. METHODS Six consecutive patients with moderate to severe AD who had failed standard treatment were treated with tofacitinib citrate. Response to treatment was assessed using the Scoring of AD index. RESULTS Decreased body surface area involvement of dermatitis and decreased erythema, edema/papulation, lichenification, and excoriation were observed in all patients. The Scoring of AD index decreased by 66.6% from 36.5 to 12.2 (P < .05) during 8 to 29 weeks of treatment. There were no adverse events. LIMITATIONS Small sample size, lack of placebo control group, and the possibility of bias are limitations. CONCLUSION The oral Janus kinase inhibitor tofacitinib citrate may be beneficial in the treatment of moderate to severe AD.
Collapse
Affiliation(s)
- Lauren L Levy
- Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut
| | - Jennifer Urban
- Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut
| | - Brett A King
- Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut.
| |
Collapse
|
28
|
Glosson NL, Bruns HA, Kaplan MH. Wheezing and itching: The requirement for STAT proteins in allergic inflammation. JAKSTAT 2014; 1:3-12. [PMID: 24058746 PMCID: PMC3670132 DOI: 10.4161/jkst.19086] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 12/16/2011] [Indexed: 12/16/2022] Open
Abstract
The development of allergic inflammation requires the orchestration of gene expression from the inflamed tissue and from the infiltrating immune cells. Since many of the cytokines that promote allergic inflammation signal through hematopoietin family receptors, the Signal Transducer and Activator of Transcription (STAT) family have obligate roles in pro-allergic cytokine-induced gene regulation in multiple cell types. In this review, we summarize work defining the contribution of each of the STAT family members to the development of allergic inflammation, using data from mouse models of allergic inflammation, studies on patient samples and correlations with single nucleotide polymorphisms in STAT genes.
Collapse
Affiliation(s)
- Nicole L Glosson
- Department of Pediatrics; Herman B. Wells Center for Pediatric Research; Department of Microbiology and Immunology; Indiana University School of Medicine; Indianapolis, IN USA
| | | | | |
Collapse
|
29
|
Bao L, Zhang H, Chan LS. The involvement of the JAK-STAT signaling pathway in chronic inflammatory skin disease atopic dermatitis. JAKSTAT 2013; 2:e24137. [PMID: 24069552 PMCID: PMC3772104 DOI: 10.4161/jkst.24137] [Citation(s) in RCA: 163] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Revised: 02/26/2013] [Accepted: 02/27/2013] [Indexed: 12/18/2022] Open
Abstract
Atopic dermatitis (AD), a common chronic inflammatory skin disease, is characterized by inflammatory cell skin infiltration. The JAK-STAT pathway has been shown to play an essential role in the dysregulation of immune responses in AD, including the exaggeration of Th2 cell response, the activation of eosinophils, the maturation of B cells, and the suppression of regulatory T cells (Tregs). In addition, the JAK-STAT pathway, activated by IL-4, also plays a critical role in the pathogenesis of AD by upregulating epidermal chemokines, pro-inflammatroy cytokines, and pro-angiogenic factors as well as by downregulating antimicrobial peptides (AMPs) and factors responsible for skin barrier function. In this review, we will highlight the recent advances in our understanding of the JAK-STAT pathway in the pathogenesis of AD.
Collapse
Affiliation(s)
- Lei Bao
- Department of Dermatology; University of Illinois; Chicago, IL USA
| | | | | |
Collapse
|
30
|
Qiu R, Yang Y, Zhao H, Li J, Xin Q, Shan S, Liu Y, Dang J, Yu X, Gong Y, Liu Q. Signal transducer and activator of transcription 6 directly regulates human ORMDL3 expression. FEBS J 2013; 280:2014-26. [PMID: 23461825 DOI: 10.1111/febs.12225] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 02/23/2013] [Accepted: 02/28/2013] [Indexed: 12/23/2022]
Abstract
Orosomucoid-like 3 (ORMDL3) has been associated with asthma and a series of autoimmune disorders, and is involved in endoplasmic reticulum-mediated inflammatory responses. However, its clinical significance and the molecular mechanism underlying its expression are still largely unclear. To elucidate the mechanisms of human ORMDL3 transcriptional regulation, we cloned a 1.5 kb genomic DNA fragment containing the putative promoter region and evaluated its transcriptional activity in a luciferase reporter system by deletion analysis. We identified a 68 bp region that functions as a minimal promoter. Bioinformatics analysis predicted that the -64 to -56 bp region contained a signal transducer and activator of transcription 6 (STAT6) binding site. Electrophoretic mobility shift assay and chromatin immunoprecipitation demonstrated that STAT6 bound to its binding site within the ORMDL3 promoter. STAT6 over-expression or knockdown trans-activated or trans-inhibited, respectively, the ORMDL3 promoter containing the STAT6-binding motif. Treatment with interleukins 4 or 13 increased ORMDL3 promoter activity as well as endogenous ORMDL3 expression. Immunoprecipitation and ChIP/Re-ChIP assays revealed that STAT6 and p300 exist in the same protein complex that binds to the ORMDL3 promoter. Our study confirmed that STAT6 plays important roles in regulating the expression of human ORMDL3 by directly binding to the promoter region, which may shed light on a possible role in various human diseases.
Collapse
Affiliation(s)
- Rongfang Qiu
- Department of Medical Genetics and Key Laboratory for Experimental Teratology of the Ministry of Education, Shandong University School of Medicine, Jinan, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Chen H, Jiang Z. The essential adaptors of innate immune signaling. Protein Cell 2012; 4:27-39. [PMID: 22996173 PMCID: PMC4875439 DOI: 10.1007/s13238-012-2063-0] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 07/22/2012] [Indexed: 12/20/2022] Open
Abstract
Microbial components and the endogenous molecules released from damaged cells can stimulate germ-line-encoded pattern recognition receptors (PRRs) to transduce signals to the hub of the innate immune signaling network-the adaptor proteins MyD88/TRIF/MAVS/STING/Caspase-1, where integrated signals relay to the relevant transcription factors IRF3/IRF7/NF-κB/ AP-1 and the signal transducer and activator of transcription 6 (STAT6) to trigger the expression of type I interferons and inflammatory cytokines or the assembly of inflammasomes. Most pleiotropic cytokines are secreted and bind to specific receptors, activating the signaling pathways including JAK-STAT for the proliferation, differentiation and functional capacity of immune cells. This review focuses on several critical adaptors in innate immune signaling cascades and recent progress in their molecular mechanisms.
Collapse
Affiliation(s)
- Huihui Chen
- School of Life Sciences, Peking University, Beijing, 100871 China
| | - Zhengfan Jiang
- School of Life Sciences, Peking University, Beijing, 100871 China
| |
Collapse
|
32
|
Mehrotra P, Hollenbeck A, Riley JP, Li F, Patel RJ, Akhtar N, Goenka S. Poly (ADP-ribose) polymerase 14 and its enzyme activity regulates T(H)2 differentiation and allergic airway disease. J Allergy Clin Immunol 2012; 131:521-31.e1-12. [PMID: 22841009 DOI: 10.1016/j.jaci.2012.06.015] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 06/07/2012] [Accepted: 06/07/2012] [Indexed: 11/24/2022]
Abstract
BACKGROUND IL-4 and signal transducer and activator of transcription 6 (STAT6) play an important role in the progression of allergic airway disease (AAD) or asthma. IL-4 and STAT6 mediate T(H)2 responses in T cells and immunoglobulin class-switching to IgE in B cells. Both T(H)2 responses and IgE promote the asthmatic condition. We have previously demonstrated that poly (ADP-ribose) polymerase (PARP) 14, a member of the PARP family of proteins, regulates the transcription function of STAT6. However, the role of PARP-14 in AAD is not known. OBJECTIVE Here we investigate the role of PARP-14 and the enzyme activity associated with it in a model of AAD dependent on airway hyperresponsiveness and lung inflammation. We also elucidate the mechanism by which PARP-14 regulates AAD. METHODS The role of PARP-14 and its enzyme activity in AAD and T(H)2 differentiation were examined by using a murine model of AAD and in vitro T(H) cell differentiation. RESULTS PARP-14-deficient animals show reduced lung pathology and IgE levels when compared with control animals. Treating mice with a pharmacologic inhibitor for PARP activity reduced the severity of airway hyperresponsiveness and lung inflammation. Mechanistically, our data indicate that PARP-14 and its enzyme activity aid in the differentiation of T cells toward a T(H)2 phenotype by regulating the binding of STAT6 to the Gata3 promoter. CONCLUSION PARP-14 and the catalytic activity associated with it promote T(H)2 differentiation and AAD in a murine model, and targeting PARP-14 might be a potential new therapy for allergic asthma.
Collapse
Affiliation(s)
- Purvi Mehrotra
- HB Wells Center for Pediatric Research and the Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Ind, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Signal transducer and activator of transcription (STAT) proteins are critical mediators of cytokine signaling. Among the seven STAT proteins, STAT6 is activated by IL-4 and IL-13 and plays a predominant role in the immune system. However, there is increasing evidence that STAT6 may function in other tissues and organ systems. IL-4, IL-13, and STAT6 promote humoral immunity, clearance of helminthic parasites as well as the pathogenesis of allergic disorders like asthma, food allergies, and atopic dermatitis. In this review, we will describe our current understanding of the biological functions of STAT6 and summarize recent advances in understanding the molecular mechanisms by which STAT6 regulates transcription.
Collapse
Affiliation(s)
- Shreevrat Goenka
- HB Wells Center of Pediatric Research, Department of Pediatrics, Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, 46202, USA.
| | | |
Collapse
|
34
|
Stritesky GL, Muthukrishnan R, Sehra S, Goswami R, Pham D, Travers J, Nguyen ET, Levy DE, Kaplan MH. The transcription factor STAT3 is required for T helper 2 cell development. Immunity 2011; 34:39-49. [PMID: 21215659 DOI: 10.1016/j.immuni.2010.12.013] [Citation(s) in RCA: 169] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 10/18/2010] [Accepted: 11/04/2010] [Indexed: 12/18/2022]
Abstract
Signal transducer and activator of transcription (STAT) family members direct the differentiation of T helper cells, with specific STAT proteins promoting distinct effector subsets. STAT6 is required for the development of T helper 2 (Th2) cells, whereas STAT3 promotes differentiation of Th17 and follicular helper T cell subsets. We demonstrated that STAT3 was also activated during Th2 cell development and was required for the expression of Th2 cell-associated cytokines and transcription factors. STAT3 bound directly to Th2 cell-associated gene loci and was required for the ability of STAT6 to bind target genes. In vivo, STAT3 deficiency in T cells eliminated the allergic inflammation in mice sensitized and challenged with ovalbumin or transgenic for constitutively active STAT6. Thus, STAT3 cooperates with STAT6 in promoting Th2 cell development. These results demonstrate that differentiating T helper cells integrate multiple STAT protein signals during Th2 cell development.
Collapse
Affiliation(s)
- Gretta L Stritesky
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Dong L, Zhang X, Fu X, Zhang X, Gao X, Zhu M, Wang X, Yang Z, Jensen ON, Saarikettu J, Yao Z, Silvennoinen O, Yang J. PTB-associated splicing factor (PSF) functions as a repressor of STAT6-mediated Ig epsilon gene transcription by recruitment of HDAC1. J Biol Chem 2010; 286:3451-9. [PMID: 21106524 DOI: 10.1074/jbc.m110.168377] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Regulation of transcription requires cooperation between sequence-specific transcription factors and numerous coregulatory proteins. In IL-4/IL-13 signaling several coactivators for STAT6 have been identified, but the molecular mechanisms of STAT6-mediated gene transcription are still not fully understood. Here we identified by proteomic approach that the PTB-associated splicing factor (PSF) interacts with STAT6. In intact cells the interaction was observed only after IL-4 stimulation. The IL-4-induced tyrosine phosphorylation of both STAT6 and PSF is a prerequisite for the efficient association of the two proteins. Functional analysis demonstrated that ectopic expression of PSF resulted in inhibition of STAT6-mediated transcriptional activation and mRNA expression of the Igε germline heavy chain gene, whereas knockdown of PSF increased the STAT6-mediated responses. PSF recruited histone deacetylase 1 (HDAC1) to the STAT6 transcription complex, which resulted in reduction of H3 acetylation at the promoter regions of Ig heavy chain germline Igε and inhibition of STAT6-mediated transcription. In addition, the HDACs inhibitor trichostatin A (TSA) enhanced H3 acetylation, and reverted the PSF-mediated transcriptional repression of Igε gene transcription. In summary, these results identify PSF as a repressor of STAT6-mediated transcription that functions through recruitment of HDAC to the STAT6 transcription complex, and delineates a novel regulatory mechanism of IL-4 signaling that may have implications in the pathogenesis of allergic diseases and pharmacological HDAC inhibition in lymphomas.
Collapse
Affiliation(s)
- Lijie Dong
- Department of Immunology, Basic Medical College, Tianjin Medical University, Heping District, Qixiangtai Road No. 22, Tianjin 300070, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Yao W, Chang J, Sehra S, Travers JB, Chang CH, Tepper RS, Kaplan MH. Altered cytokine production by dendritic cells from infants with atopic dermatitis. Clin Immunol 2010; 137:406-14. [PMID: 20880754 DOI: 10.1016/j.clim.2010.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Revised: 09/01/2010] [Accepted: 09/02/2010] [Indexed: 01/17/2023]
Abstract
Dendritic cells (DC) are potent initiators of immune responses, compared to other professional antigen-presenting cells, based on their ability to capture antigen, express high amounts of MHC and co-stimulatory molecules, and to secrete immunostimulatory cytokines. Altered functions of DC in atopic individuals have been observed, though it is not clear if this is a cause or a result of the development of allergic disease. In this report we demonstrate altered cytokine production by DC isolated from infants with atopic dermatitis but without a diagnosis of asthma, compared to infants with non-atopic dermatitis. Increased production of IL-6, IL-10 and IFNα from DC isolated from atopic infants is less apparent when DC from infants were examined 1 year later. An increase in the same cytokines was observed in neonatal mice that are genetically predisposed towards allergic inflammation. These results suggest that an atopic environment promotes altered cytokine production by DC from infants.
Collapse
Affiliation(s)
- Weiguo Yao
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Regulation in chronic obstructive pulmonary disease: the role of regulatory T-cells and Th17 cells. Clin Sci (Lond) 2010; 119:75-86. [PMID: 20402669 DOI: 10.1042/cs20100033] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
COPD (chronic obstructive pulmonary disease) is an inflammatory disorder of the airways, which is associated with irreversible airway obstruction. The pathological hallmarks of COPD are destruction of the lung parenchyma (pulmonary emphysema), inflammation of the central airways (chronic bronchitis) and inflammation of the peripheral airways (respiratory bronchiolitis). Tobacco smoking is established as the main aetiological factor for COPD. A maladaptive modulation of inflammatory responses to inhalation of noxious particles and gases is generally accepted as being a key central pathogenic process; however, the precise regulatory mechanisms of the disease are poorly understood. Two cell types are known to be important in immune regulation, namely regulatory T-cells and the newly identified Th17 (T-helper 17) cells. Both types of cells are subsets of CD4 T-lymphocytes and modulate the immune response through secretion of cytokines, for example IL (interleukin)-10 and IL-17 respectively. The present review will begin by describing the current understanding of inflammatory cell involvement in the disease process, and then focus on the possible role of subsets of regulatory and helper T-cells in COPD.
Collapse
|
38
|
Sehra S, Yao Y, Howell MD, Nguyen ET, Kansas GS, Leung DYM, Travers JB, Kaplan MH. IL-4 regulates skin homeostasis and the predisposition toward allergic skin inflammation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:3186-90. [PMID: 20147633 PMCID: PMC2837507 DOI: 10.4049/jimmunol.0901860] [Citation(s) in RCA: 148] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
IL-4 promotes the development of Th2 cells and allergic inflammation. In atopic dermatitis lesions, IL-4 decreases the expression of multiple genes associated with innate defense, including genes in the epidermal differentiation complex (EDC) that regulate epidermal barrier function. However, it is not clear whether IL-4 also contributes to homeostatic control of EDC genes. In this report, we demonstrate that expression of EDC genes and barrier function is increased in the absence of endogenous IL-4. Mice that express a constitutively active Stat6 (Stat6VT) are prone to the development of allergic skin inflammation and have decreased expression of EDC genes. IL-4 deficiency protects Stat6VT transgenic mice from the development of allergic skin inflammation and decreased recovery time in barrier function following skin irritation, with a concomitant increase in EDC gene expression. These data suggest that IL-4 plays an important role in regulating epidermal homeostasis and innate barrier function.
Collapse
Affiliation(s)
- Sarita Sehra
- Department of Pediatrics, Indiana University, School of Medicine, Indianapolis, IN 46202
- Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN 46202
| | - Yongxue Yao
- Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN 46202
- Department of Dermatology, Indiana University, School of Medicine, Indianapolis, IN 46202
| | | | - Evelyn T. Nguyen
- Department of Pediatrics, Indiana University, School of Medicine, Indianapolis, IN 46202
- Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN 46202
| | - Geoffrey S. Kansas
- Department of Microbiology-Immunology, Northwestern University Medical School, Chicago IL 60611
| | | | - Jeffrey B. Travers
- Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN 46202
- Department of Dermatology, Indiana University, School of Medicine, Indianapolis, IN 46202
- L. Roudebush VA Medical Center, Indiana University, School of Medicine, Indianapolis, IN 46202
| | - Mark H. Kaplan
- Department of Pediatrics, Indiana University, School of Medicine, Indianapolis, IN 46202
- Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN 46202
| |
Collapse
|
39
|
Hu L, Edamatsu H, Takenaka N, Ikuta S, Kataoka T. Crucial role of phospholipase Cepsilon in induction of local skin inflammatory reactions in the elicitation stage of allergic contact hypersensitivity. THE JOURNAL OF IMMUNOLOGY 2009; 184:993-1002. [PMID: 20007527 DOI: 10.4049/jimmunol.0901816] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Phospholipase Cepsilon (PLCepsilon) is an effector of Ras/Rap small GTPases. We previously demonstrated that PLCepsilon plays a crucial role in development of phorbor ester-induced skin inflammation, which is intimately involved in the promotion of skin carcinogenesis. In this study, we have examined its role in local skin inflammatory reactions during development of contact hypersensitivity toward a hapten 2,4-dinitrofluorobenzene (DNFB). PLCepsilon(+/+) and PLCepsilon(-/-) mice were sensitized with DNFB, followed by a DNFB challenge on the ears. PLCepsilon(-/-) mice exhibited substantially attenuated inflammatory reactions compared with PLCepsilon(+/+) mice as shown by suppression of ear swelling, neutrophil infiltration, and proinflammatory cytokine production. In contrast, the extent and kinetics of CD4+ T cell infiltration showed no difference depending on the PLCepsilon background. Adoptive transfer of CD4+ T cells from the sensitized mice to naive mice between PLCepsilon(+/+) and PLCepsilon(-/-) backgrounds indicated that PLCepsilon exerts its function in cells other than CD4+ T cells, presumably fibroblasts or keratinocytes of the skin, to augment inflammatory reactions during the elicitation stage of contact hypersensitivity. Moreover, dermal fibroblasts and epidermal keratinocytes cultured from the skin expressed proinflammatory cytokines in a PLCepsilon-dependent manner on stimulation with T cell-derived cytokines such as IL-17, IFN-gamma, TNF-alpha, and IL-4. These results indicate that PLCepsilon plays a crucial role in induction of proinflammatory cytokine expression in fibroblasts and keratinocytes at the challenged sites, where infiltrated CD4+ T cells produce their intrinsic cytokines, thereby augmenting the local inflammatory reactions.
Collapse
Affiliation(s)
- Lizhi Hu
- Division of Molecular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | | | | | | | | |
Collapse
|
40
|
O'Malley JT, Eri RD, Stritesky GL, Mathur AN, Chang HC, Hogenesch H, Srinivasan M, Kaplan MH. STAT4 isoforms differentially regulate Th1 cytokine production and the severity of inflammatory bowel disease. THE JOURNAL OF IMMUNOLOGY 2008; 181:5062-70. [PMID: 18802110 DOI: 10.4049/jimmunol.181.7.5062] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
STAT4, a critical regulator of inflammation in vivo, can be expressed as two alternative splice forms, a full-length STAT4alpha, and a STAT4beta isoform lacking a C-terminal transactivation domain. Each isoform is sufficient to program Th1 development through both common and distinct subsets of target genes. However, the ability of these isoforms to mediate inflammation in vivo has not been examined. Using a model of colitis that develops following transfer of CD4(+) CD45RB(high) T cells expressing either the STAT4alpha or STAT4beta isoform into SCID mice, we determined that although both isoforms mediate inflammation and weight loss, STAT4beta promotes greater colonic inflammation and tissue destruction. This correlates with STAT4 isoform-dependent expression of TNF-alpha and GM-CSF in vitro and in vivo, but not Th1 expression of IFN-gamma or Th17 expression of IL-17, which were similar in STAT4alpha- and STAT4beta-expressing T cells. Thus, higher expression of a subset of inflammatory cytokines from STAT4beta-expressing T cells correlates with the ability of STAT4beta-expressing T cells to mediate more severe inflammatory disease.
Collapse
Affiliation(s)
- John T O'Malley
- Department of Pediatrics, HB Wells Center for Pediatric Research and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | |
Collapse
|