1
|
Yu J, Mei J, Zuo D, Zhang M, Yu S, Li F, Wang J, Bi D, Ma S, Wang J, Yin ZJ. Inflammatory factor-mediated miR-155/SOCS1 signaling axis leads to Treg impairment in systemic lupus erythematosus. Int Immunopharmacol 2024; 141:113013. [PMID: 39213866 DOI: 10.1016/j.intimp.2024.113013] [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: 06/20/2024] [Revised: 08/05/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is an autoimmune disorder associated with the decrease and functional impairment of regulatory T cells (Tregs). In the current study, we explored the interplay of miR-155 and suppressor of cytokine signaling 1 (SOCS1) in regulating Treg function and stability in SLE. METHODS Clinical samples from healthy subjects and SLE patients were collected, and a mouse model of SLE was established to profile the expression pattern of miR-155 and SCOS1 in Tregs. Tregs isolated from mouse spleen were stimulated by inflammatory cytokines to confirm involvement of miR-155/SOCS1 axis in dictating Treg stability and function. We also administrated synthetic miR-155 inhibitor in SLE animal model to evaluate the potential effect on rescuing Treg function and alleviating SLE progression. RESULTS Tregs from SLE patients and SLE-induced mice exhibited a downregulation of SOCS1 and an upregulation of miR-155. In Tregs stimulated by inflammatory cytokines, Nuclear factor kappa B (NF-κB) signaling activation was required for the change of SOCS1 and miR-155 expression. miR-155 served as a negative regulator to dampen SOCS1 expression in inflammation-stimulated Tregs. The transfection of miR-155 mimic impaired the suppressive function and differentiation of Tregs through targeting SOCS1. In contrast, miR-155 inhibition improved Treg function under inflammatory stimulation and alleviated SLE conditions in the mouse model. CONCLUSION Inflammation-induced miR-155 impairs Treg stability and function in SLE through decreasing SOCS1 expression. Targeting miR-155 might be developed as an intervention to mitigate SLE conditions.
Collapse
Affiliation(s)
- Juan Yu
- Department of Rheumatology, The First People's Hospital of Yunnan Province, The Affliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
| | - Jian Mei
- Department of Rheumatology, The First People's Hospital of Yunnan Province, The Affliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
| | - Dachen Zuo
- Department of Rheumatology, The First People's Hospital of Yunnan Province, The Affliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
| | - Mingxing Zhang
- Department of Rheumatology, The First People's Hospital of Yunnan Province, The Affliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
| | - Shengnan Yu
- Department of Rheumatology, The First People's Hospital of Yunnan Province, The Affliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
| | - Fayou Li
- Department of Rheumatology, The First People's Hospital of Yunnan Province, The Affliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
| | - Juan Wang
- Department of Rheumatology, The First People's Hospital of Yunnan Province, The Affliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
| | - Danyan Bi
- Department of Rheumatology, The First People's Hospital of Yunnan Province, The Affliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
| | - Sha Ma
- Department of Rheumatology, The First People's Hospital of Yunnan Province, The Affliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
| | - Jing Wang
- Department of Rheumatology, The First People's Hospital of Yunnan Province, The Affliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
| | - Zi-Jing Yin
- Department of Rheumatology, The First People's Hospital of Yunnan Province, The Affliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China.
| |
Collapse
|
2
|
Leifer VP, Fang F, Song L, Kim J, Papanikolaou JF, Smeeton J, Thomopoulos S. Single-cell RNA-sequencing analysis of immune and mesenchymal cell crosstalk in the developing enthesis. Sci Rep 2024; 14:26839. [PMID: 39500962 PMCID: PMC11538517 DOI: 10.1038/s41598-024-77958-7] [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: 06/29/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
Autoimmunity underlies many painful disorders, such as enthesopathies, which localize to the enthesis. From infiltration of the synovium and axial skeleton by B cells, to disturbances in the ratio of M1/M2 enthesis macrophages, to CD8 + T cell mediated inflammation, autoimmune dysregulation is becoming increasingly well characterized in enthesopathies. Tissue resident B cells, macrophages, neutrophils, and T cells have also been localized in healthy human entheses. However, the potential developmental origins, presence, and role of immune cells (ICs) in enthesis development is not known. Here, we use single-cell RNA-sequencing analysis to describe IC subtypes present in the enthesis before, during, and after mineralization, and to infer regulatory interactions between ICs and mesenchymal cells (MCs). We report the presence of nine phenotypically distinct IC subtypes, including B cells, macrophages, neutrophils, and T cells. We find that specific IC subtypes may promote MC-proliferation and differentiation, and that MCs may regulate IC phenotype and autoimmunity. Our findings suggest that bidirectional regulatory interactions between ICs and MCs may be important to enthesis mineralization, and suggest that progenitor MCs have a unique ability to limit autoimmunity during development.
Collapse
Affiliation(s)
- Valia P Leifer
- Department of Orthopedic Surgery, Columbia University, New York, NY, 10032, USA
| | - Fei Fang
- Department Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Lee Song
- Department of Orthopedic Surgery, Columbia University, New York, NY, 10032, USA
| | - Jieon Kim
- Department of Orthopedic Surgery, Columbia University, New York, NY, 10032, USA
| | - John F Papanikolaou
- Department of Orthopedic Surgery, Columbia University, New York, NY, 10032, USA
| | - Joanna Smeeton
- Department of Rehabilitation and Regenerative Medicine, Columbia Stem Cell Initiative, Columbia University, New York, NY, 10032, USA
- Department of Genetics and Development, Columbia Stem Cell Initiative, Columbia University, New York, NY, 10032, USA
| | - Stavros Thomopoulos
- Department of Orthopedic Surgery, Columbia University, New York, NY, 10032, USA.
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA.
| |
Collapse
|
3
|
Michel M, Crickx E, Fattizzo B, Barcellini W. Autoimmune haemolytic anaemias. Nat Rev Dis Primers 2024; 10:82. [PMID: 39487134 DOI: 10.1038/s41572-024-00566-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/27/2024] [Indexed: 11/04/2024]
Abstract
Adult autoimmune haemolytic anaemias (AIHAs) include different subtypes of a rare autoimmune disease in which autoantibodies targeting autoantigens expressed on the membrane of autologous red blood cells (RBCs) are produced, leading to their accelerated destruction. In the presence of haemolytic anaemia, the direct antiglobulin test is the cornerstone of AIHA diagnosis. AIHAs are classified according to the isotype and the thermal optimum of the autoantibody into warm (wAIHAs), cold and mixed AIHAs. wAIHAs, the most frequent type of AIHAs, are associated with underlying conditions in ~50% of cases. In wAIHA, IgG autoantibody reacts with autologous RBCs at 37 °C, leading to antibody-dependent cell-mediated cytotoxicity and increased phagocytosis of RBCs in the spleen. Cold AIHAs include cold agglutinin disease (CAD) and cold agglutinin syndrome (CAS) when there is an underlying condition. CAD and cold agglutinin syndrome are IgM cold antibody-driven AIHAs characterized by classical complement pathway-mediated haemolysis. The management of wAIHAs has long been based around corticosteroids and splenectomy and on symptomatic measures and non-specific cytotoxic agents for CAD. Rituximab and the development of complement inhibitors, such as the anti-C1s antibody sutimlimab, have changed the therapeutic landscape of AIHAs, and new promising targeted therapies are under investigation.
Collapse
Affiliation(s)
- Marc Michel
- Department of Internal Medicine and Clinical Immunology, National Reference Centre for Adult Immune Cytopenias, Henri Mondor University Hospital, Assistance Publique Hôpitaux de Paris, Université Paris-Est Créteil, Créteil, France.
| | - Etienne Crickx
- Department of Internal Medicine and Clinical Immunology, National Reference Centre for Adult Immune Cytopenias, Henri Mondor University Hospital, Assistance Publique Hôpitaux de Paris, Université Paris-Est Créteil, Créteil, France
| | - Bruno Fattizzo
- Hematology Unit, Fondazione IRCCS Ca Granda Ospedale Maggiore Policlinico, Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Wilma Barcellini
- Hematology Unit, Fondazione IRCCS Ca Granda Ospedale Maggiore Policlinico, Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
4
|
Zhang C, Liang D, Liu Z. Primary immunodeficiency as a cause of immune-mediated kidney diseases. Nephrol Dial Transplant 2024; 39:1772-1784. [PMID: 38772735 PMCID: PMC11522874 DOI: 10.1093/ndt/gfae117] [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: 01/26/2024] [Indexed: 05/23/2024] Open
Abstract
Primary immunodeficiency (PID) is no longer defined by infections alone, and autoimmunity is an accompanying manifestation of PID. Recurrent infections may trigger autoimmunity through molecular mimicry, bystander activation or superantigens. The diagnosis of PID is still challenging, but genetic analysis reveals the underlying link between PID and autoimmunity. Mutations in relevant genes affecting central and peripheral immune tolerance, regulatory T-cell function, expansion of autoreactive lymphocytes, antigen clearance, hyperactivation of type I interferon and nuclear factor-κB pathways have all been implicated in triggering autoimmunity in PID. Autoimmunity in PID leads to chronic inflammation, tissue damage and organ failure, and increases the mortality of patients with PID. The kidneys are inextricably linked with the immune system, and kidney diseases can be mediated by both infection and autoimmunity/inflammation in PID patients. The manifestations of kidney involvement in PID patients are very heterogeneous and include lupus nephritis, C3 glomerulopathy, kidney thrombotic microangiopathy, vasculitis and interstitial nephritis. Patients with PID-caused kidney diseases have defined immune function defects and may benefit from pathway-based biologics, stem cell transplantation or gene therapy. Early diagnosis and appropriate treatment of PID are crucial for reducing the mortality rate and improving organ function and quality of life.
Collapse
Affiliation(s)
- Changming Zhang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Dandan Liang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhihong Liu
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
5
|
Lamas A, Faria R, Marinho A, Vasconcelos C. The mosaic of systemic lupus erythematosus: From autoimmunity to autoinflammation and immunodeficiency and back. Autoimmun Rev 2024; 23:103675. [PMID: 39481623 DOI: 10.1016/j.autrev.2024.103675] [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: 07/20/2024] [Revised: 09/22/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024]
Abstract
The concept of an "immunological continuum model," introduced by McGonagle and McDermott in 2006, redefines the traditional dichotomy between autoimmunity and autoinflammation, proposing a spectrum where innate and adaptive immune dysregulation can co-occur, reflecting a more nuanced understanding of immune disorders. Systemic lupus erythematosus (SLE) exemplifies the complexity of this continuum, often displaying manifestations of autoimmunity, autoinflammation, and immunodeficiency. The interplay between genetic, epigenetic, hormonal, psychological, and environmental factors contributes to its distinctive immunopathological signatures. Historically recognized as a systemic disease with diverse clinical manifestations, SLE is primarily a polygenic autoimmune condition but can, however, present in monogenic forms. Examining SLE through the lens of the immunological continuum model allows for emphasis on the contributions of both innate and adaptive immunity. SLE and primary immunodeficiencies share genetic susceptibilities and clinical manifestations. Additionally, autoinflammatory mechanisms, such as inflammasome activation and interferonopathies, can play a role in SLE pathogenesis, illustrating the disease's position at the crossroads of immune dysregulation. Recognizing the diverse clinical expressions of SLE and its mimickers is critical for accurate diagnosis and targeted therapy. In conclusion, the immunological continuum model provides a comprehensive framework for understanding SLE, acknowledging its multifaceted nature and guiding future research and clinical practice toward more effective and individualized treatments. After the Mosaic of Autoimmunity, it is now the time to focus and attempt to solve the intricate mosaic of SLE.
Collapse
Affiliation(s)
- António Lamas
- Unidade de Imunologia Clínica - Unidade Local de Saúde de Santo António, Porto, Portugal; UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Portugal.
| | - Raquel Faria
- Unidade de Imunologia Clínica - Unidade Local de Saúde de Santo António, Porto, Portugal; UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Portugal
| | - António Marinho
- Unidade de Imunologia Clínica - Unidade Local de Saúde de Santo António, Porto, Portugal; UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Portugal
| | - Carlos Vasconcelos
- Unidade de Imunologia Clínica - Unidade Local de Saúde de Santo António, Porto, Portugal; UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Portugal
| |
Collapse
|
6
|
Chen J, Li M, Shang S, Cheng L, Tang Z, Huang C. LncRNA XIST/miR-381-3P/STAT1 axis as a potential biomarker for lupus nephritis. Lupus 2024; 33:1176-1191. [PMID: 39126180 DOI: 10.1177/09612033241273072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2024]
Abstract
OBJECTIVE We aim to investigate the potential roles of key genes in the development of lupus nephritis (LN), screen key biomarkers, and construct the lncRNA XIST/miR-381-3P/STAT1 axis by using bioinformatic prediction combined with clinical validation, thereby providing new targets and insights for clinical research. METHODS Gene expression microarrays GSE157293 and GSE112943 were downloaded from the GEO database to obtain differentially expressed genes (DEGs), followed by enrichment analyses on these DEGs, which were enriched and analyzed to construct a protein-protein interaction (PPI) network to screen core genes. The lncRNA-miRNA-mRNA regulatory network was predicted and constructed based on the miRNA database. 37 female patients with systemic lupus erythematosus (SLE) were recruited to validate the bioinformatics results by exploring the diagnostic value of the target ceRNA axis in LN by dual luciferase and real-time fluorescence quantitative PCR (RT-qPCR) and receiver operating characteristic (ROC). RESULTS The data represented that a total of 133 differential genes were screened in the GSE157293 dataset and 2869 differential genes in the GSE112943 dataset, yielding a total of 26 differentially co-expressed genes. Six core genes (STAT1, OAS2, OAS3, IFI44, DDX60, and IFI44L) were screened. Biological functional analysis identified key relevant pathways in LN. ROC curve analysis suggested that lncRNA XIST, miR-381-3P, and STAT1 could be used as potential molecular markers to assist in the diagnosis of LN. CONCLUSION STAT1 is a key gene in the development of LN. In conclusion, lncRNA XIST, miR-381-3P, and STAT1 can be used as new molecular markers to assist in the diagnosis of LN, and the lncRNA XIST/miR-381-3P/STAT1 axis may be a potential therapeutic target for LN.
Collapse
Affiliation(s)
- Junjie Chen
- The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Ming Li
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Shuangshuang Shang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Lili Cheng
- The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Zhongfu Tang
- The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Chuanbing Huang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
7
|
Jeanpierre M, Cognard J, Tusseau M, Riller Q, Bui LC, Berthelet J, Laurent A, Crickx E, Parlato M, Stolzenberg MC, Suarez F, Leverger G, Aladjidi N, Collardeau-Frachon S, Pietrement C, Malphettes M, Froissart A, Bole-Feysot C, Cagnard N, Rodrigues Lima F, Walzer T, Rieux-Laucat F, Belot A, Mathieu AL. Haploinsufficiency in PTPN2 leads to early-onset systemic autoimmunity from Evans syndrome to lupus. J Exp Med 2024; 221:e20232337. [PMID: 39028869 PMCID: PMC11259789 DOI: 10.1084/jem.20232337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 05/17/2024] [Accepted: 06/26/2024] [Indexed: 07/21/2024] Open
Abstract
An exome sequencing strategy employed to identify pathogenic variants in patients with pediatric-onset systemic lupus or Evans syndrome resulted in the discovery of six novel monoallelic mutations in PTPN2. PTPN2 is a phosphatase that acts as an essential negative regulator of the JAK/STAT pathways. All mutations led to a loss of PTPN2 regulatory function as evidenced by in vitro assays and by hyperproliferation of patients' T cells. Furthermore, patients exhibited high serum levels of inflammatory cytokines, mimicking the profile observed in individuals with gain-of-function mutations in STAT factors. Flow cytometry analysis of patients' blood cells revealed typical alterations associated with autoimmunity and all patients presented with autoantibodies. These findings further supported the notion that a loss of function in negative regulators of cytokine pathways can lead to a broad spectrum of autoimmune manifestations and that PTPN2 along with SOCS1 haploinsufficiency constitute a new group of monogenic autoimmune diseases that can benefit from targeted therapy.
Collapse
Affiliation(s)
- Marie Jeanpierre
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, Université Paris Cité, INSERM UMR 1163, Paris, France, IHU-Imagine, Université de Paris, Paris, France
| | - Jade Cognard
- Centre International de Recherche en Infectiologie, Inserm, U1111, CNRS, UMR5308, École Normale Supérieure de Lyon, Lyon, France
| | - Maud Tusseau
- Centre International de Recherche en Infectiologie, Inserm, U1111, CNRS, UMR5308, École Normale Supérieure de Lyon, Lyon, France
- Department of Medical Genetics, Hospices Civils de Lyon, Bron, France
| | - Quentin Riller
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, Université Paris Cité, INSERM UMR 1163, Paris, France, IHU-Imagine, Université de Paris, Paris, France
| | - Linh-Chi Bui
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Jérémy Berthelet
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, Paris, France
| | - Audrey Laurent
- National Referee Centre for Pediatric-Onset Rheumatism and Autoimmune Diseases, Hospices Civils de Lyon, Pediatric Nephrology, Rheumatology, Dermatology Unit, Mother and Children University Hospital; Lyon, France
| | - Etienne Crickx
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, Université Paris Cité, INSERM UMR 1163, Paris, France, IHU-Imagine, Université de Paris, Paris, France
- Service de Médecine Interne, Centre National de Référence des Cytopénies Auto-immunes de L’adulte, Hôpital Henri Mondor, Fédération Hospitalo-Universitaire TRUE InnovaTive TheRapy for ImmUne disordErs, Assistance Publique Hôpitaux de Paris, Université Paris Est Créteil, Créteil, France
| | - Marianna Parlato
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, Université Paris Cité, INSERM UMR 1163, Paris, France, IHU-Imagine, Université de Paris, Paris, France
| | - Marie-Claude Stolzenberg
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, Université Paris Cité, INSERM UMR 1163, Paris, France, IHU-Imagine, Université de Paris, Paris, France
| | - Felipe Suarez
- Department of Adult Hematology, Necker-Enfants Malades University Hospital and Centre de Référence des déficits Immunitaires Héréditaires, Assistance Publique Hôpitaux de Paris, INSERM U1163, Imagine Institute, Université Paris Cité, Paris, France
| | - Guy Leverger
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, UMR_S938, Assistance Publique Hôpitaux de Paris, Groupe Hospitalier Sorbonne Université, Hôpital Armand Trousseau, Paris, France
| | - Nathalie Aladjidi
- Centre de Référence National des Cytopénies Auto-immunes de l’Enfant, Bordeaux, France
- Pediatric Oncology Hemato-Immunology Unit, University Hospital, Plurithématique Centre d’Investigation Clinique, 1401, INSERM, Bordeaux, France
| | - Sophie Collardeau-Frachon
- Institute of Pathology, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Société Française de Foetopathologie Paris, Paris, France
| | - Christine Pietrement
- Centre Hospitalier Universitaire de Reims, Service de Pédiatrie Spécialisée et Généralisée, Université Reims Champagne Ardenne, Reims, France
| | - Marion Malphettes
- Service d’Immunopathologie Clinique, Saint Louis Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Antoine Froissart
- Service Médecine Interne, Hôpital Intercommunal de Créteil, Créteil, France
| | - Christine Bole-Feysot
- Genomic Platform, INSERM UMR 1163, Imagine Institute, University Paris Cité, Paris, France
| | - Nicolas Cagnard
- Bioinformatic Platform, INSERM UMR 1163, Imagine Institute, University Paris Cité, Paris, France
| | | | - Thierry Walzer
- Centre International de Recherche en Infectiologie, Inserm, U1111, CNRS, UMR5308, École Normale Supérieure de Lyon, Lyon, France
| | - Frédéric Rieux-Laucat
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, Université Paris Cité, INSERM UMR 1163, Paris, France, IHU-Imagine, Université de Paris, Paris, France
| | - Alexandre Belot
- Centre International de Recherche en Infectiologie, Inserm, U1111, CNRS, UMR5308, École Normale Supérieure de Lyon, Lyon, France
- National Referee Centre for Pediatric-Onset Rheumatism and Autoimmune Diseases, Hospices Civils de Lyon, Pediatric Nephrology, Rheumatology, Dermatology Unit, Mother and Children University Hospital; Lyon, France
| | - Anne-Laure Mathieu
- Centre International de Recherche en Infectiologie, Inserm, U1111, CNRS, UMR5308, École Normale Supérieure de Lyon, Lyon, France
| |
Collapse
|
8
|
Tobin JM, Cooper MA. PTPN2 deficiency: Amping up JAK/STAT. J Exp Med 2024; 221:e20240980. [PMID: 39028870 PMCID: PMC11259788 DOI: 10.1084/jem.20240980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2024] Open
Abstract
Identification of monogenic causes of immune dysregulation provides insight into human immune response and signaling pathways associated with autoimmunity. Here, Jeanpierre et al. (https://doi.org/10.1084/jem.20232337) identify new germline variants in the gene encoding PTPN2 associated with loss of regulatory function, enhanced JAK/STAT signaling, and early-onset autoimmunity.
Collapse
Affiliation(s)
- Joshua M. Tobin
- Division of Rheumatology/Immunology, Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA
| | - Megan A. Cooper
- Division of Rheumatology/Immunology, Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
9
|
Thuner J, Cognard J, Belot A. How to treat monogenic SLE? Best Pract Res Clin Rheumatol 2024; 38:101962. [PMID: 38876818 DOI: 10.1016/j.berh.2024.101962] [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: 04/01/2024] [Revised: 05/12/2024] [Accepted: 05/29/2024] [Indexed: 06/16/2024]
Abstract
Systemic lupus erythematosus is a rare and life-threatening autoimmune disease characterized by autoantibodies against double-stranded DNA, with an immunopathology that remains partially unclear. New insights into the disease have been provided by the discovery of key mutations leading to the development of monogenic SLE, occurring in the context of early-onset disease, syndromic lupus, or familial clustering. The increased frequency of discovering these mutations in recent years, thanks to the advent of genetic screening, has greatly enhanced our understanding of the immunopathogenesis of SLE. These monogenic defects include defective clearance of apoptotic bodies, abnormalities in nucleic acid sensing, activation of the type-I interferon pathway, and the breakdown of tolerance through B or T cell activation or lymphocyte proliferation due to anomalies in TLR signalling and/or NFκB pathway overactivation. The translation of genetic discoveries into therapeutic strategies is presented here, within the framework of personalized therapy.
Collapse
Affiliation(s)
- Jonathan Thuner
- Internal Medicine Department, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre-Bénite, France; CIRI, Centre International de Recherche en Infectiologie/International Center for Infectiology Research, Université de Lyon, INSERM, Institut National de La Santé Et de La Recherche Médicale, U1111, Université Claude Bernard Lyon 1, Lyon, France
| | - Jade Cognard
- Pediatric Rheumatology, Nephrology, Dermatology Department, CMR RAISE, Women-Mother-Child Hospital, Hospices Civils de Lyon, Bron, France; CIRI, Centre International de Recherche en Infectiologie/International Center for Infectiology Research, Université de Lyon, INSERM, Institut National de La Santé Et de La Recherche Médicale, U1111, Université Claude Bernard Lyon 1, Lyon, France
| | - Alexandre Belot
- Pediatric Rheumatology, Nephrology, Dermatology Department, CMR RAISE, Women-Mother-Child Hospital, Hospices Civils de Lyon, Bron, France; CIRI, Centre International de Recherche en Infectiologie/International Center for Infectiology Research, Université de Lyon, INSERM, Institut National de La Santé Et de La Recherche Médicale, U1111, Université Claude Bernard Lyon 1, Lyon, France; CNRS, Centre National de La Recherche Scientifique, UMR5308, Lyon, France.
| |
Collapse
|
10
|
Khan K, Zahid M, Ali N, Attaullah S, Ullah M, Khan K, Muhammad I, Abusharha A, Aschner M, Khan H. STAT3 single-nucleotide variants in autoimmune thyroid disease in the Pakhtun population of Khyber Pakhtunkhwa, Pakistan. GENE REPORTS 2024; 36:101950. [PMID: 39385969 PMCID: PMC11463997 DOI: 10.1016/j.genrep.2024.101950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
The current study was conducted to assess the relationship between the STAT3 gene variants rs744166 and rs2293152 and autoimmune thyroid disorder in the Pakhtun population of the province, of Khyber Pakhtunkhwa, Pakistan. Blood was collected from 100 healthy individuals and 400 thyroid-disordered patients. Of these, one hundred were diagnosed with Hashimoto's thyroiditis (HT), while 32 were confirmed as Grave's disease (GD) patients. T3, T4, and TSH serum levels were checked to diagnose thyroid disorders. The blood was analyzed for anti-thyroid peroxidase antibodies (Anti-TPOAb) (AESKULISA- ATPO - elisa kit), (Germany), and thyroid stimulating hormone receptor antibodies (TSHRAb), TSHR Ab elisa kit (Diametra Italy), respectively. PCR was used to amplify the targeted STAT3 gene polymorphisms from rs744166 (301 bp) and rs2293152 (365 bp) sequences and then digested by specific restriction endonucleases (AluI) and AciI respectively. The disease displayed a female predominance. The genotype TC and CC of rs744166 showed a significant relationship with Grave's disease (p = 0.002, OR = 0.28, 95 % CI = 0.11-0.77) in patients. The C allele contributed significantly to the disease in GD patients. The SNP rs2293152 significantly differed between GD patients and control (p = 0.032, OR = 0.29, 95 % CI = 0.09-0.86). Similarly, the G and C alleles showed a significant (p = 0.02) difference between GD patients and the control. No significant association was found for both SNPs in Hashimoto's thyroiditis disease. It is concluded that the STAT3 gene (rs744166 and rs2293152) was found to have a potential role in autoimmunity in GD patients. Still, it needs further studies with larger sample sizes in the Pakhtun population to understand this relationship.
Collapse
Affiliation(s)
- Khayyam Khan
- Department of Zoology, Islamia College University, 25120 Peshawar, Pakistan
| | - Muhammad Zahid
- Department of Zoology, Islamia College University, 25120 Peshawar, Pakistan
| | - Niaz Ali
- Institute of Basic Medical Sciences Khyber Medical University, 25120 Peshawar, Pakistan
| | - Sobia Attaullah
- Department of Zoology, Islamia College University, 25120 Peshawar, Pakistan
| | - Mujeeb Ullah
- Department of Zoology, Islamia College University, 25120 Peshawar, Pakistan
| | - Khalid Khan
- Department of Zoology, Islamia College University, 25120 Peshawar, Pakistan
| | - Ijaz Muhammad
- Department of Zoology, Abdul Wali Khan University Mardan, 23200 Mardan, Pakistan
| | - Ali Abusharha
- Optometry Department, Applied Medical Sciences Collage, King Saud University, P. O. Box 145111, Riyadh, Saudi Arabia
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States of America
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, 23200 Mardan, Pakistan
| |
Collapse
|
11
|
Sutanto H, Safira A, Fetarayani D. From tumor to tolerance: A comprehensive review of immune checkpoint inhibitors and immune-related adverse events. Asia Pac Allergy 2024; 14:124-138. [PMID: 39220570 PMCID: PMC11365684 DOI: 10.5415/apallergy.0000000000000146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 05/02/2024] [Indexed: 09/04/2024] Open
Abstract
The advent of immune checkpoint inhibitors (ICIs) has revolutionized the treatment landscape for various malignancies by harnessing the body's immune system to target cancer cells. However, their widespread use has unveiled a spectrum of immune-related adverse events, highlighting a critical balance between antitumor immunity and autoimmunity. This review article delves into the molecular immunology of ICIs, mapping the journey from their therapeutic action to the unintended induction of immune-related adverse events. We provide a comprehensive overview of all available ICIs, including cytotoxic T-lymphocyte-associated protein 4, programmed cell death protein 1, programmed death-ligand 1 inhibitors, and emerging targets, discussing their mechanisms of action, clinical applications, and the molecular underpinnings of associated immune-related adverse events. Special attention is given to the activation of autoreactive T cells, B cells, cytokine release, and the inflammatory cascade, which together contribute to the development of immune-related adverse events. Through a molecular lens, we explore the clinical manifestations of immune-related adverse events across organ systems, offering insights into diagnosis, management, and strategies to mitigate these adverse effects. The review underscores the importance of understanding the delicate interplay between enhancing antitumor responses and minimizing immune-related adverse events, aiming to guide future research and the development of next-generation ICIs with improved drug safety profiles.
Collapse
Affiliation(s)
- Henry Sutanto
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Ardea Safira
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Deasy Fetarayani
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
12
|
Kandoor A, Martinez G, Hitchcock JM, Angel S, Campbell L, Rizvi S, Naegle KM. CoDIAC: A comprehensive approach for interaction analysis reveals novel insights into SH2 domain function and regulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.18.604100. [PMID: 39091881 PMCID: PMC11291013 DOI: 10.1101/2024.07.18.604100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Protein domains are conserved structural and functional units and are the functional building blocks of proteins. Evolutionary expansion means that domain families are often represented by many members in a species, which are found in various configurations with other domains, which have evolved new specificity for interacting partners. Here, we develop a structure-based interface analysis to comprehensively map domain interfaces from available experimental and predicted structures, including interfaces with other macromolecules and intraprotein interfaces (such as might exist between domains in a protein). We hypothesized that a comprehensive approach to contact mapping of domains could yield new insights. Specifically, we use it to gain information about how domains selectivity interact with ligands, whether domain-domain interfaces of repeated domain partnerships are conserved across diverse proteins, and identify regions of conserved post-translational modifications, using relationship to interaction interfaces as a method to hypothesize the effect of post-translational modifications (and mutations). We applied this approach to the human SH2 domain family, an extensive modular unit that is the foundation of phosphotyrosine-mediated signaling, where we identified a novel approach to understanding the binding selectivity of SH2 domains and evidence that there is coordinated and conserved regulation of multiple SH2 domain binding interfaces by tyrosine and serine/threonine phosphorylation and acetylation, suggesting that multiple signaling systems can regulate protein activity and SH2 domain interactions in a regulated manner. We provide the extensive features of the human SH2 domain family and this modular approach, as an open source Python package for COmprehensive Domain Interface Analysis of Contacts (CoDIAC).
Collapse
Affiliation(s)
- Alekhya Kandoor
- Department of Biomedical Engineering and the Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Gabrielle Martinez
- Department of Biomedical Engineering and the Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Julianna M Hitchcock
- Department of Biomedical Engineering and the Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Savannah Angel
- Department of Biomedical Engineering and the Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Logan Campbell
- Department of Biomedical Engineering and the Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Saqib Rizvi
- Department of Biomedical Engineering and the Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Kristen M Naegle
- Department of Biomedical Engineering and the Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
13
|
Tusseau M, Belot A. [Rare Autoimmune Diseases Role of Genetics - Example of Systemic Lupus Erythematosus]. Biol Aujourdhui 2024; 218:9-18. [PMID: 39007772 DOI: 10.1051/jbio/2024005] [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: 04/30/2024] [Indexed: 07/16/2024]
Abstract
Systemic lupus erythematosus (SLE) presents a complex clinical landscape with diverse manifestations, suggesting a multifactorial etiology. However, the identification of rare monogenic forms of the disease has shed light on specific genetic defects underlying SLE pathogenesis, offering valuable insights into its underlying mechanisms and clinical heterogeneity. By categorizing these monogenic forms based on the implicated signaling pathways, such as apoptotic body clearance, type I interferon signaling, JAK-STAT pathway dysregulation, innate immune receptor dysfunction and lymphocytic abnormalities, a more nuanced understanding of SLE's molecular basis emerges. Particularly in pediatric populations, where monogenic forms are more prevalent, routine genetic testing becomes increasingly important, with a diagnostic yield of approximately 10% depending on the demographic and methodological factors involved. This approach not only enhances diagnostic accuracy but also informs personalized treatment strategies tailored to the specific molecular defects driving the disease phenotype.
Collapse
Affiliation(s)
- Maud Tusseau
- Laboratoire de génétique des cancers et maladies multifactorielles, Service de génétique médicale, Hospices Civils de Lyon, Bron, France - Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard, Lyon 1, CNRS UMR5308, ENS de Lyon, Lyon, France
| | - Alexandre Belot
- Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard, Lyon 1, CNRS UMR5308, ENS de Lyon, Lyon, France - Centre de référence des maladies rhumatologiques inflammatoires, des maladies auto-immunes et interféronopathies systémiques de l'enfant, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Bron, France - Service de néphrologie, rhumatologie, dermatologie pédiatrique, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
14
|
Zhou Y, Song HM. Type I interferon pathway in pediatric systemic lupus erythematosus. World J Pediatr 2024; 20:653-668. [PMID: 38914753 PMCID: PMC11269505 DOI: 10.1007/s12519-024-00811-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/27/2024] [Indexed: 06/26/2024]
Abstract
BACKGROUND The role of type I interferon (IFN-I) signaling in systemic lupus erythematosus (SLE) has been well established. However, unanswered questions remain regarding the applicability of these findings to pediatric-onset SLE. The aim of this review is to provide an overview of the novel discoveries on IFN-I signaling in pediatric-onset SLE. DATA SOURCES A literature search was conducted in the PubMed database using the following keywords: "pediatric systemic lupus erythematosus" and "type I interferon". RESULTS IFN-I signaling is increased in pediatric SLE, largely due to the presence of plasmacytoid dendritic cells and pathways such as cyclic GMP-AMP synthase-stimulator of interferon genes-TANK-binding kinase 1 and Toll-like receptor (TLR)4/TLR9. Neutrophil extracellular traps and oxidative DNA damage further stimulate IFN-I production. Genetic variants in IFN-I-related genes, such as IFN-regulatory factor 5 and tyrosine kinase 2, are linked to SLE susceptibility in pediatric patients. In addition, type I interferonopathies, characterized by sustained IFN-I activation, can mimic SLE symptoms and are thus important to distinguish. Studies on interferonopathies also contribute to exploring the pathogenesis of SLE. Measuring IFN-I activation is crucial for SLE diagnosis and stratification. Both IFN-stimulated gene expression and serum IFN-α2 levels are common indicators. Flow cytometry markers such as CD169 and galectin-9 are promising alternatives. Anti-IFN therapies, such as sifalimumab and anifrolumab, show promise in adult patients with SLE, but their efficacy in pediatric patients requires further investigation. Janus kinase inhibitors are another treatment option for severe pediatric SLE patients. CONCLUSIONS This review presents an overview of the IFN-I pathway in pediatric SLE. Understanding the intricate relationship between IFN-I and pediatric SLE may help to identify potential diagnostic markers and targeted therapies, paving the way for improved patient care and outcomes.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
| | - Hong-Mei Song
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China.
| |
Collapse
|
15
|
Caldirola MS, Daiana E, Gomez Raccio AC, García AL, Bernacchia A, Medín M, Gaillard MI, Di Giovanni D. Case Report: Common variable immunodeficiency phenotype and granulomatous-lymphocytic interstitial lung disease with a novel SOCS1 variant. Front Pediatr 2024; 12:1423858. [PMID: 39005503 PMCID: PMC11239428 DOI: 10.3389/fped.2024.1423858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 06/10/2024] [Indexed: 07/16/2024] Open
Abstract
Common variable immunodeficiency is a heterogeneous symptomatic group of inborn errors of immunity that mainly affects antibodies production and/or function, predisposing patients to recurrent and severe infections. More than half of them usually develop autoimmunity, lymphoproliferation, enteropathy, and malignancies. Among these conditions, chronic lung disease such as granulomatous-lymphocytic interstitial lung disease is one of the leading causes of death in these patients. Recently, many genes that play a key role in B and T cells' development, maintenance, and/or cytokines signaling pathways have been implicated in the pathogenesis of the disease. Here, we describe the first Argentinian patient presenting with common variable immunodeficiency and granulomatous-lymphocytic interstitial lung disease, harboring two in cis heterozygous variants in the SOCS1 gene.
Collapse
Affiliation(s)
- María Soledad Caldirola
- Servicio de Inmunología, Hospital de Niños “Dr. Ricardo Gutiérrez”, Buenos Aires, Argentina
- Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas (IMIPP—CONICET-GCBA), Buenos Aires, Argentina
| | - Espantoso Daiana
- Servicio de Inmunología, Hospital de Niños “Dr. Ricardo Gutiérrez”, Buenos Aires, Argentina
| | | | - Ana Luz García
- Servicio de Inmunología, Hospital de Niños “Dr. Ricardo Gutiérrez”, Buenos Aires, Argentina
| | - Agustin Bernacchia
- Servicio de Inmunología, Hospital de Niños “Dr. Ricardo Gutiérrez”, Buenos Aires, Argentina
| | - Martín Medín
- Servicio de Anatomía Patológica, Hospital de Niños “Dr. Ricardo Gutiérrez”, Buenos Aires, Argentina
| | - Maria Isabel Gaillard
- Servicio de Inmunología, Hospital de Niños “Dr. Ricardo Gutiérrez”, Buenos Aires, Argentina
| | - Daniela Di Giovanni
- Servicio de Inmunología, Hospital de Niños “Dr. Ricardo Gutiérrez”, Buenos Aires, Argentina
| |
Collapse
|
16
|
Bidgood GM, Keating N, Doggett K, Nicholson SE. SOCS1 is a critical checkpoint in immune homeostasis, inflammation and tumor immunity. Front Immunol 2024; 15:1419951. [PMID: 38947335 PMCID: PMC11211259 DOI: 10.3389/fimmu.2024.1419951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 05/28/2024] [Indexed: 07/02/2024] Open
Abstract
The Suppressor of Cytokine Signaling (SOCS) family proteins are important negative regulators of cytokine signaling. SOCS1 is the prototypical member of the SOCS family and functions in a classic negative-feedback loop to inhibit signaling in response to interferon, interleukin-12 and interleukin-2 family cytokines. These cytokines have a critical role in orchestrating our immune defence against viral pathogens and cancer. The ability of SOCS1 to limit cytokine signaling positions it as an important immune checkpoint, as evidenced by the detection of detrimental SOCS1 variants in patients with cytokine-driven inflammatory and autoimmune disease. SOCS1 has also emerged as a key checkpoint that restricts anti-tumor immunity, playing both a tumor intrinsic role and impacting the ability of various immune cells to mount an effective anti-tumor response. In this review, we describe the mechanism of SOCS1 action, focusing on the role of SOCS1 in autoimmunity and cancer, and discuss the potential for new SOCS1-directed cancer therapies that could be used to enhance adoptive immunotherapy and immune checkpoint blockade.
Collapse
Affiliation(s)
- Grace M. Bidgood
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Narelle Keating
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Karen Doggett
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Sandra E. Nicholson
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
17
|
Carpenter S, O'Neill LAJ. From periphery to center stage: 50 years of advancements in innate immunity. Cell 2024; 187:2030-2051. [PMID: 38670064 PMCID: PMC11060700 DOI: 10.1016/j.cell.2024.03.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/24/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024]
Abstract
Over the past 50 years in the field of immunology, something of a Copernican revolution has happened. For a long time, immunologists were mainly concerned with what is termed adaptive immunity, which involves the exquisitely specific activities of lymphocytes. But the other arm of immunity, so-called "innate immunity," had been neglected. To celebrate Cell's 50th anniversary, we have put together a review of the processes and components of innate immunity and trace the seminal contributions leading to the modern state of this field. Innate immunity has joined adaptive immunity in the center of interest for all those who study the body's defenses, as well as homeostasis and pathology. We are now entering the era where therapeutic targeting of innate immune receptors and downstream signals hold substantial promise for infectious and inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Susan Carpenter
- University of California Santa Cruz, 1156 High St., Santa Cruz, CA 95064, USA.
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
18
|
Körholz J, Chen LS, Strauss T, Schuetz C, Dalpke AH. One gene to rule them all - clinical perspectives of a potent suppressor of cytokine signaling - SOCS1. Front Immunol 2024; 15:1385190. [PMID: 38711523 PMCID: PMC11070515 DOI: 10.3389/fimmu.2024.1385190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 03/25/2024] [Indexed: 05/08/2024] Open
Abstract
The discovery of Suppressor of Cytokine Signaling 1 (SOCS1) in 1997 marked a significant milestone in understanding the regulation of Janus kinase/Signal transducer and activator of transcription (JAK/STAT) signaling pathways. Subsequent research deciphered its cellular functions, and recent insights into SOCS1 deficiencies in humans underscored its critical role in immune regulation. In humans, SOCS-haploinsufficiency (SOCS1-HI) presents a diverse clinical spectrum, encompassing autoimmune diseases, infection susceptibility, and cancer. Variability in disease manifestation, even within families sharing the same genetic variant, raises questions about clinical penetrance and the need for individualized treatments. Current therapeutic strategies include JAK inhibition, with promising results in controlling inflammation in SOCS1-HI patients. Hematopoietic stem cell transplantation and gene therapy emerge as promising avenues for curative treatments. The evolving landscape of SOCS1 research, emphasizes the need for a nuanced understanding of genetic variants and their functional consequences.
Collapse
Affiliation(s)
- Julia Körholz
- Department of Pediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- University Center for Chronic Immunodeficiencies (UCID), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Lan-Sun Chen
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Medical Faculty, University Heidelberg, Heidelberg, Germany
- University Hospital Heidelberg, Heidelberg, Germany
| | - Timmy Strauss
- Department of Pediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- University Center for Chronic Immunodeficiencies (UCID), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- University Center for Rare Diseases, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Catharina Schuetz
- Department of Pediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- University Center for Chronic Immunodeficiencies (UCID), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- University Center for Rare Diseases, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Alexander H. Dalpke
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Medical Faculty, University Heidelberg, Heidelberg, Germany
- University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
19
|
Granel J, Fernandes H, Bader-Meunier B, Guth A, Richer O, Pillet P, Leverger G, Ducassou S, Fahd M, Pasquet M, Garnier N, Barlogis V, Guitton C, Jeziorski E, Thomas C, Bayart S, Cheikh N, Paillard C, Abou Chahla W, Chastagner P, Neven B, Millot F, Lejeune J, Li-Thiao Te V, Armari-Alla C, Briandet C, Carausu L, Deparis M, Piguet C, Benadiba J, Marie-Cardine A, Stephan JL, Pellier I, Pluchart C, Doré E, Michaux K, Héritier S, Leblanc T, Aladjidi N. Antinuclear antibody-associated autoimmune cytopenia in childhood is a risk factor for systemic lupus erythematosus. Blood 2024; 143:1576-1585. [PMID: 38227934 DOI: 10.1182/blood.2023021884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 12/12/2023] [Accepted: 12/15/2023] [Indexed: 01/18/2024] Open
Abstract
ABSTRACT Autoimmune cytopenia (AIC) in children may be associated with positive antinuclear antibodies (ANA) and may progress to systemic lupus erythematosus (SLE). We evaluated the risk of progression to SLE of childhood-onset ANA-associated AIC. In the French national prospective OBS'CEREVANCE cohort, the long-term outcome of children with ANA-associated AIC (ANA titer ≥1/160) and a subgroup of children who developed SLE were described. ANA were positive in 355 of 1803 (20%) children with AIC. With a median follow-up of 5.8 (range, 0.1-29.6) years, 79 of 355 (22%) patients developed SLE at a median age of 14.5 (1.1-21.4) years; 20% of chronic immune thrombocytopenic purpura, 19% of autoimmune hemolytic anemia, and 45% of Evans syndrome. None of the patients with ANA-negative test developed SLE. Severe manifestations of SLE were observed in 21 patients, and 2 patients died. In multivariate analysis including patients with positive ANA within the first 3 months after AIC diagnosis, age >10 years at AIC diagnosis (relative risk [RR], 3.67; 95% confidence interval [CI], 1.18-11.4; P = .024) and ANA titer >1/160 (RR, 5.28; 95% CI, 1.20-23.17; P = .027) were associated with the occurrence of SLE after AIC diagnosis. ANA-associated AIC is a risk factor for progression to SLE, especially in children with an initial ANA titer >1/160 and an age >10 years at AIC diagnosis. ANA screening should be recommended in children with AIC, and patients with ANA should be monitored long-term for SLE, with special attention to the transition period. This trial was registered at www.ClinicalTrials.gov as #NCT05937828.
Collapse
Affiliation(s)
- Jérôme Granel
- Paediatric Clinical Immunology, Pellegrin Hospital, CIC1401, INSERM CICP, Bordeaux University Hospital, Bordeaux, France
- Centre de Référence National des Cytopénies Auto-immunes de l'Enfant, Bordeaux, France
| | - Helder Fernandes
- Paediatric Clinical Immunology, Pellegrin Hospital, CIC1401, INSERM CICP, Bordeaux University Hospital, Bordeaux, France
| | - Brigitte Bader-Meunier
- Paediatric Haematology-Immunology and Rheumatology Department, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Amandine Guth
- Paediatric Department, Pontarlier Hospital, Pontarlier, France
| | - Olivier Richer
- Paediatric Clinical Immunology, Pellegrin Hospital, CIC1401, INSERM CICP, Bordeaux University Hospital, Bordeaux, France
| | - Pascal Pillet
- Paediatric Clinical Immunology, Pellegrin Hospital, CIC1401, INSERM CICP, Bordeaux University Hospital, Bordeaux, France
| | - Guy Leverger
- Centre de Référence National des Cytopénies Auto-immunes de l'Enfant, Bordeaux, France
- Paediatric Oncology Immunology Haematology Unit, Armand-Trousseau University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Stéphane Ducassou
- Paediatric Clinical Immunology, Pellegrin Hospital, CIC1401, INSERM CICP, Bordeaux University Hospital, Bordeaux, France
- Centre de Référence National des Cytopénies Auto-immunes de l'Enfant, Bordeaux, France
| | - Mony Fahd
- Centre de Référence National des Cytopénies Auto-immunes de l'Enfant, Bordeaux, France
- Paediatric Haematology and Immunology Unit, Robert-Debré University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Marlène Pasquet
- Paediatric Oncology Immunology Haematology Unit, Children's University Hospital, Toulouse, France
| | - Nathalie Garnier
- Institute of Paediatric Haematology and Oncology, Hospices Civils de Lyon, Lyon, France
| | - Vincent Barlogis
- Department of Paediatric Haematology, La Timone Hospital, Marseille University Hospital, Marseille, France
| | - Corinne Guitton
- Department of Paediatrics, Bicêtre University Hospital, Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France
| | - Eric Jeziorski
- Paediatric Oncology Haematology Unit, Arnaud de Villeneuve University Hospital, Montpellier, France
| | - Caroline Thomas
- Paediatric Haematology Unit, Nantes University Hospital, Nantes, France
| | - Sophie Bayart
- Paediatric Haematology Unit, Rennes University Hospital, Rennes, France
| | - Nathalie Cheikh
- Department of Paediatric Haematology-Oncology, Besançon University Hospital, Besançon, France
| | - Catherine Paillard
- Department of Paediatric Haematology and Oncology, Hautepierre University Hospital, Strasbourg, France
| | - Wadih Abou Chahla
- Department of Paediatric Haematology, Jeanne de Flandre Hospital, Lille University Hospital, Lille, France
| | - Pascal Chastagner
- Department of Paediatric Haematology and Oncology, Children's University Hospital, Nancy, France
| | - Bénédicte Neven
- Paediatric Haematology-Immunology and Rheumatology Department, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Frédéric Millot
- Department of Paediatric Haematology, Poitiers University Hospital, Poitiers, France
| | - Julien Lejeune
- Department of Paediatric Haematology-Oncology, Clocheville Hospital, Tours University Hospital, Tours, France
| | - Valérie Li-Thiao Te
- Department of Paediatric Haematology/Oncology, Amiens University Hospital, Amiens, France
| | - Corinne Armari-Alla
- Paediatric Haematology-Oncology Department, Grenoble University Hospital, Grenoble, France
| | - Claire Briandet
- Department of Paediatrics, Dijon University Hospital, Dijon, France
| | - Liana Carausu
- Department of Paediatric Hematology, CHU de Brest, Brest, France
| | - Marianna Deparis
- Paediatric Oncology- Haematology Unit Department, Caen University Hospital, Caen, France
| | - Christophe Piguet
- Paediatric Oncology Hematology Unit, Limoges University Hospital, Limoges, France
| | - Joy Benadiba
- Department of Haematology-Oncology Paediatrics, Nice University Hospital, Nice, France
| | - Aude Marie-Cardine
- Department of Paediatric Haematology and Oncology, Rouen University Hospital, Rouen, France
| | - Jean-Louis Stephan
- University Hospital of Saint Etienne, North Hospital, Department of Paediatric Oncology, Saint Etienne, France
| | | | - Claire Pluchart
- Paediatric Haematology-Oncology Unit, Institut Jean Godinot, Reims University Hospital, Reims, France
| | - Eric Doré
- Paediatric Unit, Clermont-Ferrand University Hospital, Clermont-Ferrand, France
| | - Katell Michaux
- Paediatric Unit, Martinique University Hospital, Fort-de-France, France
| | - Sébastien Héritier
- Centre de Référence National des Cytopénies Auto-immunes de l'Enfant, Bordeaux, France
- Paediatric Oncology Immunology Haematology Unit, Armand-Trousseau University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Thierry Leblanc
- Centre de Référence National des Cytopénies Auto-immunes de l'Enfant, Bordeaux, France
- Paediatric Haematology and Immunology Unit, Robert-Debré University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Nathalie Aladjidi
- Paediatric Clinical Immunology, Pellegrin Hospital, CIC1401, INSERM CICP, Bordeaux University Hospital, Bordeaux, France
- Centre de Référence National des Cytopénies Auto-immunes de l'Enfant, Bordeaux, France
| |
Collapse
|
20
|
Skoracka K, Hryhorowicz S, Tovoli F, Raiteri A, Rychter AM, Słomski R, Dobrowolska A, Granito A, Krela-Kaźmierczak I. From an understanding of etiopathogenesis to novel therapies-what is new in the treatment of celiac disease? Front Pharmacol 2024; 15:1378172. [PMID: 38698821 PMCID: PMC11063403 DOI: 10.3389/fphar.2024.1378172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/02/2024] [Indexed: 05/05/2024] Open
Abstract
Celiac disease, a chronic autoimmune disorder caused by genetic factors and exposure to gluten, is increasingly being recognized and diagnosed in both children and adults. Scientists have been searching for a cure for this disease for many years, but despite the impressive development of knowledge in this field, a gluten-free diet remains the only recommended therapy for all patients. At the same time, the increasing diagnosis of celiac disease in adults, which was considered a childhood disease in the 20th century, has opened a discussion on the etiopathology of the disease, which is proven to be very complex and involves genetic, immunological, nutritional, environmental and gut microbiota-related factors. In this review, we extensively discuss these factors and summarize the knowledge of the proposed state-of-the-art treatments for celiac disease to address the question of whether a better understanding of the etiopathogenesis of celiac disease has opened new directions for therapy.
Collapse
Affiliation(s)
- Kinga Skoracka
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Francesco Tovoli
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Alberto Raiteri
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Anna Maria Rychter
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
- Laboratory of Nutrigenetics, Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Ryszard Słomski
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Agnieszka Dobrowolska
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Alessandro Granito
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Iwona Krela-Kaźmierczak
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
- Laboratory of Nutrigenetics, Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
21
|
Tusseau M, Khaldi-Plassart S, Cognard J, Viel S, Khoryati L, Benezech S, Mathieu AL, Rieux-Laucat F, Bader-Meunier B, Belot A. Mendelian Causes of Autoimmunity: the Lupus Phenotype. J Clin Immunol 2024; 44:99. [PMID: 38619739 DOI: 10.1007/s10875-024-01696-8] [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: 01/26/2024] [Accepted: 03/25/2024] [Indexed: 04/16/2024]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease that is characterized by its large heterogeneity in terms of clinical presentation and severity. The pathophysiology of SLE involves an aberrant autoimmune response against various tissues, an excess of apoptotic bodies, and an overproduction of type-I interferon. The genetic contribution to the disease is supported by studies of monozygotic twins, familial clustering, and genome-wide association studies (GWAS) that have identified numerous risk loci. In the early 70s, complement deficiencies led to the description of familial forms of SLE caused by a single gene defect. High-throughput sequencing has recently identified an increasing number of monogenic defects associated with lupus, shaping the concept of monogenic lupus and enhancing our insights into immune tolerance mechanisms. Monogenic lupus (moSLE) should be suspected in patients with either early-onset lupus or syndromic lupus, in male, or in familial cases of lupus. This review discusses the genetic basis of monogenic SLE and proposes its classification based on disrupted pathways. These pathways include defects in the clearance of apoptotic cells or immune complexes, interferonopathies, JAK-STATopathies, TLRopathies, and T and B cell dysregulations.
Collapse
Affiliation(s)
- Maud Tusseau
- Centre International de Recherche en Infectiologie, Inserm, U1111, University Claude Bernard, Lyon 1, Centre National de La Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France
| | - Samira Khaldi-Plassart
- National Referee Centre for Rheumatic and AutoImmune and Systemic Diseases in Children, European Reference Network (ERN) for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) Center, Hospices Civils de Lyon, Lyon, France
- Pediatric Nephrology, Rheumatology, Dermatology Unit, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Lyon, France
| | - Jade Cognard
- Centre International de Recherche en Infectiologie, Inserm, U1111, University Claude Bernard, Lyon 1, Centre National de La Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France
- Pediatric Nephrology, Rheumatology, Dermatology Unit, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Lyon, France
| | - Sebastien Viel
- Centre International de Recherche en Infectiologie, Inserm, U1111, University Claude Bernard, Lyon 1, Centre National de La Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France
| | - Liliane Khoryati
- Centre International de Recherche en Infectiologie, Inserm, U1111, University Claude Bernard, Lyon 1, Centre National de La Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France
| | - Sarah Benezech
- Centre International de Recherche en Infectiologie, Inserm, U1111, University Claude Bernard, Lyon 1, Centre National de La Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France
| | - Anne-Laure Mathieu
- Centre International de Recherche en Infectiologie, Inserm, U1111, University Claude Bernard, Lyon 1, Centre National de La Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France
| | - Fréderic Rieux-Laucat
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, INSERM UMR 1163, Université Paris Cité, Paris, France
| | - Brigitte Bader-Meunier
- National Referee Centre for Rheumatic and AutoImmune and Systemic Diseases in Children, European Reference Network (ERN) for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) Center, Hospices Civils de Lyon, Lyon, France
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, INSERM UMR 1163, Université Paris Cité, Paris, France
- Department for Immunology, Hematology and Pediatric Rheumatology, Necker Hospital, APHP, Institut IMAGINE, Paris, France
| | - Alexandre Belot
- Centre International de Recherche en Infectiologie, Inserm, U1111, University Claude Bernard, Lyon 1, Centre National de La Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France.
- National Referee Centre for Rheumatic and AutoImmune and Systemic Diseases in Children, European Reference Network (ERN) for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) Center, Hospices Civils de Lyon, Lyon, France.
- Pediatric Nephrology, Rheumatology, Dermatology Unit, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Lyon, France.
| |
Collapse
|
22
|
Chaimowitz NS, Smith MR, Forbes Satter LR. JAK/STAT defects and immune dysregulation, and guiding therapeutic choices. Immunol Rev 2024; 322:311-328. [PMID: 38306168 DOI: 10.1111/imr.13312] [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] [Indexed: 02/03/2024]
Abstract
Inborn errors of immunity (IEIs) encompass a diverse spectrum of genetic disorders that disrupt the intricate mechanisms of the immune system, leading to a variety of clinical manifestations. Traditionally associated with an increased susceptibility to recurrent infections, IEIs have unveiled a broader clinical landscape, encompassing immune dysregulation disorders characterized by autoimmunity, severe allergy, lymphoproliferation, and even malignancy. This review delves into the intricate interplay between IEIs and the JAK-STAT signaling pathway, a critical regulator of immune homeostasis. Mutations within this pathway can lead to a wide array of clinical presentations, even within the same gene. This heterogeneity poses a significant challenge, necessitating individually tailored therapeutic approaches to effectively manage the diverse manifestations of these disorders. Additionally, JAK-STAT pathway defects can lead to simultaneous susceptibility to both infection and immune dysregulation. JAK inhibitors, with their ability to suppress JAK-STAT signaling, have emerged as powerful tools in controlling immune dysregulation. However, questions remain regarding the optimal selection and dosing regimens for each specific condition. Hematopoietic stem cell transplantation (HSCT) holds promise as a curative therapy for many JAK-STAT pathway disorders, but this procedure carries significant risks. The use of JAK inhibitors as a bridge to HSCT has been proposed as a potential strategy to mitigate these risks.
Collapse
Affiliation(s)
- Natalia S Chaimowitz
- Department of Immunology, Cook Children's Medical Center, Fort Worth, Texas, USA
| | - Madison R Smith
- UT Health Sciences Center McGovern Medical School, Houston, Texas, USA
| | - Lisa R Forbes Satter
- Department of Pediatrics, Division of Immunology, Allergy and Retrovirology, Baylor College of Medicine, Houston, Texas, USA
- William T. Shearer Texas Children's Hospital Center for Human Immunobiology, Houston, Texas, USA
| |
Collapse
|
23
|
Baumgartner F, Bamopoulos SA, Faletti L, Hsiao HJ, Holz M, Gonzalez-Menendez I, Solé-Boldo L, Horne A, Gosavi S, Özerdem C, Singh N, Liebig S, Ramamoorthy S, Lehmann M, Demel U, Kühl AA, Wartewig T, Ruland J, Wunderlich FT, Schick M, Walther W, Rose-John S, Haas S, Quintanilla-Martinez L, Feske S, Ehl S, Glauben R, Keller U. Activation of gp130 signaling in T cells drives T H17-mediated multi-organ autoimmunity. Sci Signal 2024; 17:eadc9662. [PMID: 38377177 DOI: 10.1126/scisignal.adc9662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 01/31/2024] [Indexed: 02/22/2024]
Abstract
The IL-6-gp130-STAT3 signaling axis is a major regulator of inflammation. Activating mutations in the gene encoding gp130 and germline gain-of-function mutations in STAT3 (STAT3GOF) are associated with multi-organ autoimmunity, severe morbidity, and adverse prognosis. To dissect crucial cellular subsets and disease biology involved in activated gp130 signaling, the gp130-JAK-STAT3 axis was constitutively activated using a transgene, L-gp130, specifically targeted to T cells. Activating gp130 signaling in T cells in vivo resulted in fatal, early onset, multi-organ autoimmunity in mice that resembled human STAT3GOF disease. Female mice had more rapid disease progression than male mice. On a cellular level, gp130 signaling induced the activation and effector cell differentiation of T cells, promoted the expansion of T helper type 17 (TH17) cells, and impaired the activity of regulatory T cells. Transcriptomic profiling of CD4+ and CD8+ T cells from these mice revealed commonly dysregulated genes and a gene signature that, when applied to human transcriptomic data, improved the segregation of patients with transcriptionally diverse STAT3GOF mutations from healthy controls. The findings demonstrate that increased gp130-STAT3 signaling leads to TH17-driven autoimmunity that phenotypically resembles human STAT3GOF disease.
Collapse
Affiliation(s)
- Francis Baumgartner
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité (Junior) (Digital) Clinician Scientist Program, 10178 Berlin, Germany
- German Cancer Consortium (DKTK), partner site Berlin, a partnership between DKFZ and Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Stefanos A Bamopoulos
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité (Junior) (Digital) Clinician Scientist Program, 10178 Berlin, Germany
- German Cancer Consortium (DKTK), partner site Berlin, a partnership between DKFZ and Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Laura Faletti
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, University Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Hsiang-Jung Hsiao
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Maximilian Holz
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- German Cancer Consortium (DKTK), partner site Berlin, a partnership between DKFZ and Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Irene Gonzalez-Menendez
- Institute of Pathology and Neuropathology, Comprehensive Cancer Center, Eberhard Karls University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," Eberhard Karls University, 72076 Tübingen, Germany
- German Cancer Consortium (DKTK), partner site Tübingen, a partnership between DKFZ and Eberhard Karls University of Tübingen, 72076 Tübingen, Germany
| | - Llorenç Solé-Boldo
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany
| | - Arik Horne
- German Cancer Consortium (DKTK), partner site Berlin, a partnership between DKFZ and Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sanket Gosavi
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Ceren Özerdem
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany
| | - Nikita Singh
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Sven Liebig
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Senthilkumar Ramamoorthy
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, University Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg, 79110 Freiburg, Germany
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Malte Lehmann
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- iPATH.Berlin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Benjamin Franklin, 12203 Berlin, Germany
| | - Uta Demel
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité (Junior) (Digital) Clinician Scientist Program, 10178 Berlin, Germany
| | - Anja A Kühl
- iPATH.Berlin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Benjamin Franklin, 12203 Berlin, Germany
| | - Tim Wartewig
- Institute for Clinical Chemistry and Pathobiochemistry, Technische Universität München, 81675 Munich, Germany
- Center of Molecular and Cellular Oncology, Yale School of Medicine, Yale University, New Haven, CT 06510, USA
| | - Jürgen Ruland
- Institute for Clinical Chemistry and Pathobiochemistry, Technische Universität München, 81675 Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and Technische Universität München, 81675 Munich, Germany
| | - Frank T Wunderlich
- Obesity and Cancer, Max Planck Institute for Metabolism Research, 50931 Cologne, Germany
| | - Markus Schick
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- German Cancer Consortium (DKTK), partner site Berlin, a partnership between DKFZ and Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Wolfgang Walther
- Experimental and Clinical Research Center, Charité Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Robert-Rössle Str. 10, 13125 Berlin, Germany
- EPO GmbH Berlin-Buch, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-Universität zu Kiel, 24118 Kiel, Germany
| | - Simon Haas
- German Cancer Consortium (DKTK), partner site Berlin, a partnership between DKFZ and Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany
- Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ) and DKFZ - ZMBH Alliance, 69120 Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany
| | - Leticia Quintanilla-Martinez
- Institute of Pathology and Neuropathology, Comprehensive Cancer Center, Eberhard Karls University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," Eberhard Karls University, 72076 Tübingen, Germany
- German Cancer Consortium (DKTK), partner site Tübingen, a partnership between DKFZ and Eberhard Karls University of Tübingen, 72076 Tübingen, Germany
| | - Stefan Feske
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Stephan Ehl
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, University Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Rainer Glauben
- German Cancer Consortium (DKTK), partner site Berlin, a partnership between DKFZ and Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Ulrich Keller
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- German Cancer Consortium (DKTK), partner site Berlin, a partnership between DKFZ and Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| |
Collapse
|
24
|
Ilangumaran S, Gui Y, Shukla A, Ramanathan S. SOCS1 expression in cancer cells: potential roles in promoting antitumor immunity. Front Immunol 2024; 15:1362224. [PMID: 38415248 PMCID: PMC10897024 DOI: 10.3389/fimmu.2024.1362224] [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: 12/27/2023] [Accepted: 01/31/2024] [Indexed: 02/29/2024] Open
Abstract
Suppressor of cytokine signaling 1 (SOCS1) is a potent regulator immune cell responses and a proven tumor suppressor. Inhibition of SOCS1 in T cells can boost antitumor immunity, whereas its loss in tumor cells increases tumor aggressivity. Investigations into the tumor suppression mechanisms so far focused on tumor cell-intrinsic functions of SOCS1. However, it is possible that SOCS1 expression in tumor cells also regulate antitumor immune responses in a cell-extrinsic manner via direct and indirect mechanisms. Here, we discuss the evidence supporting the latter, and its implications for antitumor immunity.
Collapse
Affiliation(s)
- Subburaj Ilangumaran
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | | | | |
Collapse
|
25
|
Bonelli M, Kerschbaumer A, Kastrati K, Ghoreschi K, Gadina M, Heinz LX, Smolen JS, Aletaha D, O'Shea J, Laurence A. Selectivity, efficacy and safety of JAKinibs: new evidence for a still evolving story. Ann Rheum Dis 2024; 83:139-160. [PMID: 37923366 PMCID: PMC10850682 DOI: 10.1136/ard-2023-223850] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/18/2023] [Indexed: 11/07/2023]
Abstract
Fundamental insight gained over the last decades led to the discovery of cytokines as pivotal drivers of inflammatory diseases such as rheumatoid arthritis, psoriasis/psoriasis arthritis, inflammatory bowel diseases, atopic dermatitis and spondylarthritis. A deeper understanding of the pro-inflammatory and anti-inflammatory effects of various cytokines has prompted new cytokine-targeting therapies, which revolutionised the treatment options in the last years for patients with inflammatory disorders. Disease-associated immune responses typically involve a complex interplay of multiple cytokines. Therefore, blockade of one single cytokine does not necessarily lead to a persistent remission in all patients with inflammatory disorders and fostered new therapeutic strategies targeting intracellular pathways shared by multiple cytokines. By inhibiting JAK-STAT signalling pathways common to families of cytokines, JAK-inhibitors (JAKinibs) have created a new paradigm for the treatment of inflammatory diseases. Multiple agents have been approved for various disorders and more are being investigated for several new indications. Second-generation selective JAKinibs have been devised with the aim to achieve an increased selectivity and a possible reduced risk of side effects. In the current review, we will summarise the current body of evidence of pan versus selective JAKinibs and the most recent insights on new side effects and indications, including COVID-19.
Collapse
Affiliation(s)
- Michael Bonelli
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Andreas Kerschbaumer
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Kastriot Kastrati
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Kamran Ghoreschi
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Massimo Gadina
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Leonhard X Heinz
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Josef S Smolen
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Daniel Aletaha
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - John O'Shea
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Arian Laurence
- Translational Gastroenterology Unit, Department of Haematology, University College Hospital, UCLH Hospitals NHS Trust, University of Oxford, Oxford, UK
| |
Collapse
|
26
|
Reuschlé Q, Van Heddegem L, Bosteels V, Moncan M, Depauw S, Wadier N, Maréchal S, De Nolf C, Delgado V, Messai Y, Stolzenberg MC, Magérus A, Werck A, Olagne J, Li Q, Lefevre G, Korganow AS, Rieux-Laucat F, Janssens S, Soulas-Sprauel P. Loss of function of XBP1 splicing activity of IRE1α favors B cell tolerance breakdown. J Autoimmun 2024; 142:103152. [PMID: 38071801 DOI: 10.1016/j.jaut.2023.103152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/27/2023] [Accepted: 11/20/2023] [Indexed: 03/23/2024]
Abstract
Anti-nuclear antibodies are the hallmark of autoimmune diseases such as systemic lupus erythematosus (SLE) and scleroderma. However, the molecular mechanisms of B cell tolerance breakdown in these pathological contexts are poorly known. The study of rare familial forms of autoimmune diseases could therefore help to better describe common biological mechanisms leading to B cell tolerance breakdown. By Whole-Exome Sequencing, we identified a new heterozygous mutation (p.R594C) in ERN1 gene, encoding IRE1α (Inositol-Requiring Enzyme 1α), in a multiplex family with several members presenting autoantibody-mediated autoimmunity. Using human cell lines and a knock-in (KI) transgenic mouse model, we showed that this mutation led to a profound defect of IRE1α ribonuclease activity on X-Box Binding Protein 1 (XBP1) splicing. The KI mice developed a broad panel of autoantibodies, however in a subclinical manner. These results suggest that a decrease of spliced form of XBP1 (XBP1s) production could contribute to B cell tolerance breakdown and give new insights into the function of IRE1α which are important to consider for the development of IRE1α targeting strategies.
Collapse
Affiliation(s)
- Quentin Reuschlé
- Laboratoire d'ImmunoRhumatologie Moléculaire, INSERM UMR_S1109, F-67000, Strasbourg, France; Strasbourg University, Faculty of Pharmacy and Faculty of Medicine, Strasbourg, France; Arthritis R&D, Neuilly sur Seine, France
| | - Laurien Van Heddegem
- Laboratory for ER Stress and Inflammation, VIB Center for Inflammation Research, Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Victor Bosteels
- Laboratory for ER Stress and Inflammation, VIB Center for Inflammation Research, Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Matthieu Moncan
- Université Paris Cité, Laboratoire d'immunogénétique des maladies auto-immunes pédiatriques, Institut Imagine, INSERM UMR_S1163, Paris, France
| | - Sabine Depauw
- Laboratoire d'ImmunoRhumatologie Moléculaire, INSERM UMR_S1109, F-67000, Strasbourg, France; Strasbourg University, Faculty of Pharmacy and Faculty of Medicine, Strasbourg, France
| | - Nadège Wadier
- Laboratoire d'ImmunoRhumatologie Moléculaire, INSERM UMR_S1109, F-67000, Strasbourg, France; Strasbourg University, Faculty of Pharmacy and Faculty of Medicine, Strasbourg, France
| | - Sandra Maréchal
- Laboratory for ER Stress and Inflammation, VIB Center for Inflammation Research, Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Clint De Nolf
- Laboratory for ER Stress and Inflammation, VIB Center for Inflammation Research, Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Barriers in Inflammation, VIB Center for Inflammation Research, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Virginia Delgado
- Laboratoire d'ImmunoRhumatologie Moléculaire, INSERM UMR_S1109, F-67000, Strasbourg, France; Strasbourg University, Faculty of Pharmacy and Faculty of Medicine, Strasbourg, France
| | | | - Marie-Claude Stolzenberg
- Université Paris Cité, Laboratoire d'immunogénétique des maladies auto-immunes pédiatriques, Institut Imagine, INSERM UMR_S1163, Paris, France
| | - Aude Magérus
- Université Paris Cité, Laboratoire d'immunogénétique des maladies auto-immunes pédiatriques, Institut Imagine, INSERM UMR_S1163, Paris, France
| | - Angélique Werck
- Department of Pathology, University Hospital, Strasbourg, France
| | - Jérôme Olagne
- Department of Pathology, University Hospital, Strasbourg, France; Department of Adult Nephrology, University Hospital, Strasbourg, France
| | - Quan Li
- Department of Immunology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Guillaume Lefevre
- Inserm, U1286 - INFINITE - Institute for Translational Research in Inflammation, University of Lille, CHU Lille, Lille, France
| | - Anne-Sophie Korganow
- Laboratoire d'ImmunoRhumatologie Moléculaire, INSERM UMR_S1109, F-67000, Strasbourg, France; Strasbourg University, Faculty of Pharmacy and Faculty of Medicine, Strasbourg, France; Department of Clinical Immunology and Internal Medicine, National Reference Center for Systemic Autoimmune Diseases (CNR RESO), Tertiary Center for Primary Immunodeficiency, Strasbourg University Hospital, F-67000, Strasbourg, France
| | - Frédéric Rieux-Laucat
- Université Paris Cité, Laboratoire d'immunogénétique des maladies auto-immunes pédiatriques, Institut Imagine, INSERM UMR_S1163, Paris, France
| | - Sophie Janssens
- Laboratory for ER Stress and Inflammation, VIB Center for Inflammation Research, Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Pauline Soulas-Sprauel
- Laboratoire d'ImmunoRhumatologie Moléculaire, INSERM UMR_S1109, F-67000, Strasbourg, France; Strasbourg University, Faculty of Pharmacy and Faculty of Medicine, Strasbourg, France; Department of Clinical Immunology and Internal Medicine, National Reference Center for Systemic Autoimmune Diseases (CNR RESO), Tertiary Center for Primary Immunodeficiency, Strasbourg University Hospital, F-67000, Strasbourg, France.
| |
Collapse
|
27
|
Gruber C, Lee A, Buta S, Khattri S, Gottlieb AB, Frost JM, Bowcock AM, Ho HE, Bogunovic D. IL4Rα and IL17A Blockade Rescue Autoinflammation in SOCS1 Haploinsufficiency. J Clin Immunol 2023; 44:36. [PMID: 38157076 PMCID: PMC11218038 DOI: 10.1007/s10875-023-01635-z] [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: 08/29/2023] [Accepted: 12/02/2023] [Indexed: 01/03/2024]
Abstract
By inhibition of JAK-STAT signaling, SOCS1 acts as a master regulator of the cytokine response across numerous tissue types and cytokine pathways. Haploinsufficiency of SOCS1 has recently emerged as a monogenic immunodysregulatory disease with marked clinical variability. Here, we describe a patient with severe dermatitis, recurrent skin infections, and psoriatic arthritis that harbors a novel heterozygous mutation in SOCS1. The variant, c.202_203delAC, generates a frameshift in SOCS1, p.Thr68fsAla*49, which leads to complete loss of protein expression. Unlike WT SOCS1, Thr68fs SOCS1 fails to inhibit JAK-STAT signaling when expressed in vitro. The peripheral immune signature from this patient was marked by a redistribution of monocyte sub-populations and hyper-responsiveness to multiple cytokines. Despite this broad hyper-response across multiple cytokine pathways in SOCS1 haploinsufficiency, the patient's clinical disease was markedly responsive to targeted IL4Rα- and IL17-blocking therapy. In accordance, the mutant allele was unable to regulate IL4Rα signaling. Further, patient cells were unresponsive to IL4/IL13 while on monoclonal antibody therapy. Together, this study reports a novel SOCS1 mutation and suggests that IL4Rα blockade may serve as an unexpected, but fruitful therapeutic target for some patients with SOCS1 haploinsufficiency.
Collapse
Affiliation(s)
- Conor Gruber
- Center for Inborn Errors of Immunity, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Angelica Lee
- Center for Inborn Errors of Immunity, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sofija Buta
- Center for Inborn Errors of Immunity, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Saakshi Khattri
- Department of Dermatology, Icahn School of Medicine at Mount Sinai Hospital, New York, NY, USA
| | - Alice B Gottlieb
- Department of Dermatology, Icahn School of Medicine at Mount Sinai Hospital, New York, NY, USA
| | - Jacqueline M Frost
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai Hospital, New York, NY, USA
| | - Anne M Bowcock
- Department of Dermatology, Icahn School of Medicine at Mount Sinai Hospital, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai Hospital, New York, NY, USA
- Department of Genetics & Genomics, Icahn School of Medicine at Mount Sinai Hospital, New York, NY, USA
| | - Hsi-En Ho
- Division of Clinical Immunology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dusan Bogunovic
- Center for Inborn Errors of Immunity, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
28
|
Mikhalkevich N, Russ E, Iordanskiy S. Cellular RNA and DNA sensing pathways are essential for the dose-dependent response of human monocytes to ionizing radiation. Front Immunol 2023; 14:1235936. [PMID: 38152396 PMCID: PMC10751912 DOI: 10.3389/fimmu.2023.1235936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 11/30/2023] [Indexed: 12/29/2023] Open
Abstract
Circulating monocytes are important players of the inflammatory response to ionizing radiation (IR). These IR-resistant immune cells migrate to radiation-damaged tissues and differentiate into macrophages that phagocytize dying cells, but also facilitate inflammation. Besides the effect of damage-associated molecular patterns, released from irradiated tissues, the inflammatory activation of monocytes and macrophages is largely dependent on IR-induced DNA damage and aberrant transcriptional activity, which may facilitate expression of type I interferons (IFN-I) and numerous inflammation-related genes. We analyzed the accumulation of dsRNA, dsDNA fragments, and RNA:DNA hybrids in the context of induction of RNA-triggered MAVS-mediated and DNA-triggered STING-mediated signaling pathways, in primary human monocytes and a monocytic cell line, THP1, in response to various doses of gamma IR. We found that exposure to lower doses (<7.5 Gy) led to the accumulation of dsRNA, along with dsDNA and RNA:DNA hybrids and activated both MAVS and STING pathway-induced gene expression and signaling activity of IFN-I. Higher doses of IR resulted in the reduced dsRNA level, degradation of RNA-sensing mediators involved in MAVS signaling and coincided with an increased accumulation of dsDNA and RNA:DNA hybrids that correlated with elevated STING signaling and NF-κB-dependent gene expression. While both pathways activate IFN-I expression, using MAVS- and STING-knockout THP1 cells, we identified differences in the spectra of interferon-stimulated genes (ISGs) that are associated with each specific signaling pathway and outlined a large group of STING signaling-associated genes. Using the RNAi technique, we found that increasing the dose of IR activates STING signaling through the DNA sensor cGAS, along with suppression of the DDX41 helicase, which is known to reduce the accumulation of RNA:DNA hybrids and thereby limit cGAS/STING signaling activity. Together, these results indicate that depending on the applied dose, IR leads to the activation of either dsRNA-induced MAVS signaling, which predominantly leads to the expression of both pro- and anti-inflammatory markers, or dsDNA-induced STING signaling that contributes to pro-inflammatory activation of the cells. While RNA:DNA hybrids boost both MAVS- and STING-mediated signaling pathways, these structures being accumulated upon high IR doses promote type I interferon expression and appear to be potent enhancers of radiation dose-dependent pro-inflammatory activation of monocytes.
Collapse
Affiliation(s)
- Natallia Mikhalkevich
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Eric Russ
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
- The American Genome Center (TAGC), Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Graduate Program of Cellular and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Sergey Iordanskiy
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Armed Forces Radiobiology Research Institute, Uniformed Services University of The Health Sciences, Bethesda, MD, United States
| |
Collapse
|
29
|
Zhang J, Lee PY, Aksentijevich I, Zhou Q. How to Build a Fire: The Genetics of Autoinflammatory Diseases. Annu Rev Genet 2023; 57:245-274. [PMID: 37562411 DOI: 10.1146/annurev-genet-030123-084224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Systemic autoinflammatory diseases (SAIDs) are a heterogeneous group of disorders caused by excess activation of the innate immune system in an antigen-independent manner. Starting with the discovery of the causal gene for familial Mediterranean fever, more than 50 monogenic SAIDs have been described. These discoveries, paired with advances in immunology and genomics, have allowed our understanding of these diseases to improve drastically in the last decade. The genetic causes of SAIDs are complex and include both germline and somatic pathogenic variants that affect various inflammatory signaling pathways. We provide an overview of the acquired SAIDs from a genetic perspective and summarize the clinical phenotypes and mechanism(s) of inflammation, aiming to provide a comprehensive understanding of the pathogenesis of autoinflammatory diseases.
Collapse
Affiliation(s)
- Jiahui Zhang
- Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Pui Y Lee
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ivona Aksentijevich
- Inflammatory Disease Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA;
| | - Qing Zhou
- Life Sciences Institute, Zhejiang University, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China;
| |
Collapse
|
30
|
Du Y, Brodeur KE, Hsu E, Chen L, Chen Q, Liu M, Cheng Q, Rosen S, Michniacki TF, Chou J, Lo MS, Platt CD, Lee PY. In cis "benign" SOCS1 variants linked to enhanced interferon signaling and autoimmunity. J Autoimmun 2023; 140:103119. [PMID: 37797401 PMCID: PMC10987394 DOI: 10.1016/j.jaut.2023.103119] [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: 07/14/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/07/2023]
Abstract
We aimed to characterize the genetic basis of disease in a family with multiple autoimmune manifestations, including systemic lupus erythematosus (SLE), immune thrombocytopenia, and autoimmune thyroiditis. Whole exome sequencing (WES) was conducted to identify candidate variants, which were analyzed by flow cytometry, immunoblotting, immunoprecipitation, and luciferase reporter assay in transfected 293T cells. Gene expression in peripheral blood mononuclear cells (PBMC) was profiled by bulk RNA sequencing and plasma cytokines were measured by proximity extension assay. In two siblings with early-onset SLE and immune thrombocytopenia, WES identified two maternally inherited in cis variants (p. Pro50Leu and p.Ala76Gly) in Suppressor of cytokine signaling 1 (SOCS1), flanking the kinase inhibitory domain that interacts with Janus kinases (JAK). Both variants were predicted to be benign by most in silico algorithms and neither alone affected the ability of SOCS1 to inhibit JAK-STAT1 signaling by functional studies. When both variants were expressed in cis, the mutant SOCS1 protein displayed decreased binding to JAK1 and reduced capacity to inhibit type I interferon (IFN-I) signaling by ∼20-30% compared to the wildtype protein. PBMC from the probands and their mother showed increased expression of interferon-inducible genes compared to healthy controls, supporting defective regulation of IFN-I signaling. Cells from all three subjects displayed heightened sensitivity to IFN-I stimulation, while response to IFN-γ, IL-4, and IL-6 was comparable to healthy controls. Our work illustrates the critical fine-tuning of IFN-I signaling by SOCS1 to prevent autoimmunity. We show that a combination of genetic variants that are individually benign may have deleterious consequences.
Collapse
Affiliation(s)
- Yan Du
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Kailey E Brodeur
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Evan Hsu
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Liang Chen
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Qian Chen
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Meng Liu
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Qi Cheng
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Seymour Rosen
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Thomas F Michniacki
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Janet Chou
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mindy S Lo
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Craig D Platt
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pui Y Lee
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
31
|
Liu M, Hsu E, Du Y, Lee PY. Suppressor of Cytokine Signaling 1 Haploinsufficiency: A New Driver of Autoimmunity and Immunodysregulation. Rheum Dis Clin North Am 2023; 49:757-772. [PMID: 37821194 DOI: 10.1016/j.rdc.2023.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Suppressor of cytokine signaling 1 (SOCS1) is a negative regulator of cytokine signaling that inhibits the activation of Janus kinases. A human disease caused by SOCS1 haploinsufficiency was first identified in 2020. To date, 18 cases of SOCS1 haploinsufficiency have been described. These patients experience enhanced activation of leukocytes and multiorgan system immunodysregulation, with immune-mediated cytopenia as the most common feature. In this review, the authors provide an overview on the biology of SOCS1 and summarize their knowledge of SOCS1 haploinsufficiency including genetics and clinical manifestations. They discuss the available treatment experience and outline an approach for the evaluation of suspected cases.
Collapse
Affiliation(s)
- Meng Liu
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA; Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Evan Hsu
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Yan Du
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA; Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Pui Y Lee
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
32
|
Mohan S. Targeted Treatment of Diseases of Immune Dysregulation. Rheum Dis Clin North Am 2023; 49:913-929. [PMID: 37821203 DOI: 10.1016/j.rdc.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Increasing molecular knowledge of autoinflammatory and autoimmune disorders has enabled more targeted treatment of these conditions. Treatment of inflammasomopathies is often aimed at interleukin-1 (IL-1) blockade, with potential use of other inhibitors targeting cytokines such as IL-18 and IL-6. Interferonopathies and some disorders with overlap features of autoimmunity and autoinflammation may improve with Janus kinase inhibition. Autoimmune conditions may also respond to inhibition of different cytokines, as well as to inhibition of T and B lymphocytes. Effective treatment is increasingly possible through targeted/precision medicine approaches.
Collapse
Affiliation(s)
- Smriti Mohan
- Division of Rheumatology, Department of Pediatrics, University of Michigan CS Mott Children's Hospital, 1500 East Medical Ctr Dr SPC 5718, Ann Arbor, MI 48109-5718, USA.
| |
Collapse
|
33
|
Pandey R, Bakay M, Hakonarson H. SOCS-JAK-STAT inhibitors and SOCS mimetics as treatment options for autoimmune uveitis, psoriasis, lupus, and autoimmune encephalitis. Front Immunol 2023; 14:1271102. [PMID: 38022642 PMCID: PMC10643230 DOI: 10.3389/fimmu.2023.1271102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/02/2023] [Indexed: 12/01/2023] Open
Abstract
Autoimmune diseases arise from atypical immune responses that attack self-tissue epitopes, and their development is intricately connected to the disruption of the JAK-STAT signaling pathway, where SOCS proteins play crucial roles. Conditions such as autoimmune uveitis, psoriasis, lupus, and autoimmune encephalitis exhibit immune system dysfunctions associated with JAK-STAT signaling dysregulation. Emerging therapeutic strategies utilize JAK-STAT inhibitors and SOCS mimetics to modulate immune responses and alleviate autoimmune manifestations. Although more research and clinical studies are required to assess their effectiveness, safety profiles, and potential for personalized therapeutic approaches in autoimmune conditions, JAK-STAT inhibitors and SOCS mimetics show promise as potential treatment options. This review explores the action, effectiveness, safety profiles, and future prospects of JAK inhibitors and SOCS mimetics as therapeutic agents for psoriasis, autoimmune uveitis, systemic lupus erythematosus, and autoimmune encephalitis. The findings underscore the importance of investigating these targeted therapies to advance treatment options for individuals suffering from autoimmune diseases.
Collapse
Affiliation(s)
- Rahul Pandey
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Marina Bakay
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pediatrics, The University of Pennsylvania School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
34
|
Kaulen LD, Denisova E, Hinz F, Hai L, Friedel D, Henegariu O, Hoffmann DC, Ito J, Kourtesakis A, Lehnert P, Doubrovinskaia S, Karschnia P, von Baumgarten L, Kessler T, Baehring JM, Brors B, Sahm F, Wick W. Integrated genetic analyses of immunodeficiency-associated Epstein-Barr virus- (EBV) positive primary CNS lymphomas. Acta Neuropathol 2023; 146:499-514. [PMID: 37495858 PMCID: PMC10412493 DOI: 10.1007/s00401-023-02613-w] [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: 05/28/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/28/2023]
Abstract
Immunodeficiency-associated primary CNS lymphoma (PCNSL) represents a distinct clinicopathological entity, which is typically Epstein-Barr virus-positive (EBV+) and carries an inferior prognosis. Genetic alterations that characterize EBV-related CNS lymphomagenesis remain unclear precluding molecular classification and targeted therapies. In this study, a comprehensive genetic analysis of 22 EBV+ PCNSL, therefore, integrated clinical and pathological information with exome and RNA sequencing (RNASeq) data. EBV+ PCNSL with germline controls carried a median of 55 protein-coding single nucleotide variants (SNVs; range 24-217) and 2 insertions/deletions (range 0-22). Genetic landscape was largely shaped by aberrant somatic hypermutation with a median of 41.01% (range 31.79-53.49%) of SNVs mapping to its target motifs. Tumors lacked established SNVs (MYD88, CD79B, PIM1) and copy number variants (CDKN2A, HLA loss) driving EBV- PCNSL. Instead, EBV+ PCNSL were characterized by SOCS1 mutations (26%), predicted to disinhibit JAK/STAT signaling, and mutually exclusive gain-of-function NOTCH pathway SNVs (26%). Copy number gains were enriched on 11q23.3, a locus directly targeted for chromosomal aberrations by EBV, that includes SIK3 known to protect from cytotoxic T-cell responses. Losses covered 5q31.2 (STING), critical for sensing viral DNA, and 17q11 (NF1). Unsupervised clustering of RNASeq data revealed two distinct transcriptional groups, that shared strong expression of CD70 and IL1R2, previously linked to tolerogenic tumor microenvironments. Correspondingly, deconvolution of bulk RNASeq data revealed elevated M2-macrophage, T-regulatory cell, mast cell and monocyte fractions in EBV+ PCNSL. In addition to novel insights into the pathobiology of EBV+ PCNSL, the data provide the rationale for the exploration of targeted therapies including JAK-, NOTCH- and CD70-directed approaches.
Collapse
Affiliation(s)
- Leon D Kaulen
- Department of Neurology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany.
- Clinical Cooperation Unit (CCU) Neuro-Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Im Neuenheimer Feld 400, 69120, Heidelberg, Germany.
| | - Evgeniya Denisova
- Division of Applied Bioinformatics, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Felix Hinz
- Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit (CCU) Neuropathology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Ling Hai
- Clinical Cooperation Unit (CCU) Neuro-Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Dennis Friedel
- Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Octavian Henegariu
- Department of Neurosurgery, Yale School of Medicine, New Haven, USA
- Department of Genetics, Yale School of Medicine, New Haven, USA
| | - Dirk C Hoffmann
- Department of Neurology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
- Clinical Cooperation Unit (CCU) Neuro-Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Jakob Ito
- Clinical Cooperation Unit (CCU) Neuro-Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Alexandros Kourtesakis
- Clinical Cooperation Unit (CCU) Neuro-Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Pascal Lehnert
- Clinical Cooperation Unit (CCU) Neuro-Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Sofia Doubrovinskaia
- Department of Neurology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Philipp Karschnia
- Department of Neurosurgery, Munich University Hospital, Ludwig Maximilians University (LMU) Munich, and German Cancer Consortium (DKTK) Partner Site, Munich, Germany
| | - Louisa von Baumgarten
- Department of Neurosurgery, Munich University Hospital, Ludwig Maximilians University (LMU) Munich, and German Cancer Consortium (DKTK) Partner Site, Munich, Germany
| | - Tobias Kessler
- Department of Neurology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
- Clinical Cooperation Unit (CCU) Neuro-Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Joachim M Baehring
- Department of Neurosurgery, Yale School of Medicine, New Haven, USA
- Department of Neurology, Yale School of Medicine, New Haven, USA
| | - Benedikt Brors
- Division of Applied Bioinformatics, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Felix Sahm
- Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany.
- Clinical Cooperation Unit (CCU) Neuropathology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.
| | - Wolfgang Wick
- Department of Neurology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany.
- Clinical Cooperation Unit (CCU) Neuro-Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Im Neuenheimer Feld 400, 69120, Heidelberg, Germany.
| |
Collapse
|
35
|
Lockwood KC, Lear TB, Rajbhandari S, McKelvey AC, Dunn SR, Boudreau ÁN, Liu Y, Chen BB. KIAA0317 regulates SOCS1 stability to ameliorate colonic inflammation. FEBS J 2023; 290:3802-3811. [PMID: 36938956 PMCID: PMC10509311 DOI: 10.1111/febs.16780] [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: 10/24/2022] [Revised: 02/20/2023] [Accepted: 03/17/2023] [Indexed: 03/21/2023]
Abstract
Dysregulated cytokine signalling is a hallmark of inflammatory bowel diseases. Inflammatory responses of the colon are regulated by the suppressor of cytokine signalling (SOCS) proteins. SOCS1 is a key member of this family, and its function is critical in maintaining an appropriate inflammatory response through the JAK/STAT signalling pathway. Dysregulation of SOCS1 protein has been identified as a causal element in colonic inflammatory diseases. Despite this, it remains unclear how SOCS1 protein is regulated. Here, we identify that SOCS1 protein is targeted for degradation by the ubiquitin proteasome system, mediated by the E3 ubiquitin ligase KIAA0317 during experimental colonic inflammation. We characterize the mechanism of protein-protein interaction and ubiquitin conjugation to SOCS1 and demonstrate that the modulation of SOCS1 protein level leads to stark effects on JAK/STAT inflammatory signalling. Together, these results provide insight into the regulation of colonic inflammation through a new mechanism of ubiquitin-based control of SOCS1 protein.
Collapse
Affiliation(s)
- Karina C. Lockwood
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA 15219, USA
| | - Travis B. Lear
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA 15219, USA
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Shristi Rajbhandari
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Alison C. McKelvey
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Sarah R. Dunn
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Áine N. Boudreau
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA 15219, USA
| | - Yuan Liu
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA 15219, USA
| | - Bill B. Chen
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA 15219, USA
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
36
|
Hale RC, Owen N, Yuan B, Chinn IK. Phenotypic Variability of SOCS1 Haploinsufficiency. J Clin Immunol 2023; 43:902-906. [PMID: 36890397 DOI: 10.1007/s10875-023-01460-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/24/2023] [Indexed: 03/10/2023]
Affiliation(s)
- Rebecca C Hale
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Nichole Owen
- Baylor College of Medicine, Baylor Genetics, Houston, TX, 77030, USA
| | - Bo Yuan
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ivan K Chinn
- Division of Immunology, Allergy and Retrovirology, Department of Pediatrics, Texas Children's Hospital and Baylor College of Medicine, 1102 Bates Ave., Ste 330, Houston, TX, 77030, USA.
| |
Collapse
|
37
|
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: 19.0] [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
|
38
|
Huang LA, Lin C, Yang L. Plumbing mysterious RNAs in "dark genome" for the conquest of human diseases. Mol Ther 2023; 31:1577-1595. [PMID: 37165619 PMCID: PMC10278048 DOI: 10.1016/j.ymthe.2023.05.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/11/2023] [Accepted: 05/05/2023] [Indexed: 05/12/2023] Open
Abstract
Next-generation sequencing has revealed that less than 2% of transcribed genes are translated into proteins, with a large portion transcribed into noncoding RNAs (ncRNAs). Among these, long noncoding RNAs (lncRNAs) represent the largest group and are pervasively transcribed throughout the genome. Dysfunctions in lncRNAs have been found in various diseases, highlighting their potential as therapeutic, diagnostic, and prognostic targets. However, challenges, such as unknown molecular mechanisms and nonspecific immune responses, and issues of drug specificity and delivery present obstacles in translating lncRNAs into clinical applications. In this review, we summarize recent publications that have explored lncRNA functions in human diseases. We also discuss challenges and future directions for developing lncRNA treatments, aiming to bridge the gap between functional studies and clinical potential and inspire further exploration in the field.
Collapse
Affiliation(s)
- Lisa A Huang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chunru Lin
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Liuqing Yang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
39
|
Toth KA, Schmitt EG, Cooper MA. Deficiencies and Dysregulation of STAT Pathways That Drive Inborn Errors of Immunity: Lessons from Patients and Mouse Models of Disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1463-1472. [PMID: 37126806 PMCID: PMC10151837 DOI: 10.4049/jimmunol.2200905] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/11/2023] [Indexed: 05/03/2023]
Abstract
The STAT family proteins provide critical signals for immune cell development, differentiation, and proinflammatory and anti-inflammatory responses. Inborn errors of immunity (IEIs) are caused by single gene defects leading to immune deficiency and/or dysregulation, and they have provided opportunities to identify genes important for regulating the human immune response. Studies of patients with IEIs due to altered STAT signaling, and mouse models of these diseases, have helped to shape current understanding of the mechanisms whereby STAT signaling and protein interactions regulate immunity. Although many STAT signaling pathways are shared, clinical and immune phenotypes in patients with monogenic defects of STAT signaling highlight both redundant and nonredundant pathways. In this review, we provide an overview of the shared and unique signaling pathways used by STATs, phenotypes of IEIs with altered STAT signaling, and recent discoveries that have provided insight into the human immune response and treatment of disease.
Collapse
Affiliation(s)
- Kelsey A. Toth
- Department of Pediatrics, Division of Rheumatology/Immunology, Washington University in St. Louis, St. Louis, MO 63110
| | - Erica G. Schmitt
- Department of Pediatrics, Division of Rheumatology/Immunology, Washington University in St. Louis, St. Louis, MO 63110
| | - Megan A. Cooper
- Department of Pediatrics, Division of Rheumatology/Immunology, Washington University in St. Louis, St. Louis, MO 63110
| |
Collapse
|
40
|
Rodari MM, Cazals-Hatem D, Uzzan M, Martin Silva N, Khiat A, Ta MC, Lhermitte L, Touzart A, Hanein S, Rouillon C, Joly F, Elmorjani A, Steffann J, Cerf-Bensussan N, Parlato M, Charbit-Henrion F. Insights into the expanding intestinal phenotypic spectrum of SOCS1 haploinsufficiency and therapeutic options. J Clin Immunol 2023:10.1007/s10875-023-01495-7. [PMID: 37156989 DOI: 10.1007/s10875-023-01495-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/17/2023] [Indexed: 05/10/2023]
Abstract
PURPOSE Hyper activation of the JAK-STAT signaling underlies the pathophysiology of many human immune-mediated diseases. Herein, the study of 2 adult patients with SOCS1 haploinsufficiency illustrates the severe and pleomorphic consequences of its impaired regulation in the intestinal tract. METHODS Two unrelated adult patients presented with gastrointestinal manifestations, one with Crohn's disease-like ileo-colic inflammation refractory to anti-TNF and the other with lymphocytic leiomyositis causing severe chronic intestinal pseudo-occlusion. Next-generation sequencing was used to identify the underlying monogenic defect. One patient received anti-IL-12/IL-23 treatment while the other received the JAK1 inhibitor, ruxolitinib. Peripheral blood, intestinal tissues, and serum samples were analyzed before-and-after JAK1 inhibitor therapy using mass cytometry, histology, transcriptomic, and Olink assay. RESULTS Novel germline loss-of-function variants in SOCS1 were identified in both patients. The patient with Crohn-like disease achieved clinical remission with anti-IL-12/IL-23 treatment. In the second patient with lymphocytic leiomyositis, ruxolitinib induced rapid resolution of the obstructive symptoms, significant decrease of the CD8+ T lymphocyte muscular infiltrate, and normalization of serum and intestinal cytokines. Decreased frequencies of circulating Treg cells, MAIT cells, and NK cells, with altered CD56bright:CD16lo:CD16hi NK subtype ratios were not modified by ruxolitinib. CONCLUSION SOCS1 haploinsufficiency can result in a broad spectrum of intestinal manifestations and need to be considered as differential diagnosis in cases of severe treatment-refractory enteropathies, including the rare condition of lymphocytic leiomyositis. This provides the rationale for genetic screening and considering JAK inhibitors in such cases.
Collapse
Affiliation(s)
- Marco M Rodari
- Université Paris-Cité, Institut Imagine, Laboratory of Intestinal Immunity, INSERM U1163, Paris, France
| | - Dominique Cazals-Hatem
- Department of Pathology, Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, Clichy, France
| | - Mathieu Uzzan
- Department of Gastroenterology, IBD unit, Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, Clichy, France
- Paris Est Créteil University UPEC, Assistance Publique-Hôpitaux de Paris (AP-HP), Henri Mondor Hospital, Gastroenterology department, Fédération Hospitalo-Universitaire TRUE InnovaTive theRapy for immUne disordErs, F-94010, Créteil, France
| | | | - Anis Khiat
- Université Paris-Cité, Institut Imagine, Laboratory of Intestinal Immunity, INSERM U1163, Paris, France
| | - Minh Chau Ta
- Department of Pathology, Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, Clichy, France
| | - Ludovic Lhermitte
- Université Paris Cité, Institut Necker Enfants-Malades INEM, Institut National de La Santé Et de La Recherche Médicale (Inserm), U1151, Paris, France
| | - Aurore Touzart
- Université Paris Cité, Institut Necker Enfants-Malades INEM, Institut National de La Santé Et de La Recherche Médicale (Inserm), U1151, Paris, France
- Laboratory of Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants-Malades 75743, Paris, France
| | - Sylvain Hanein
- Bioinformatic Platform, Institute of Genetic Diseases, INSERM UMR1163, Imagine, Université Paris-Cité and Structure Fédérative de Recherche Necker, 75015, Paris, France
| | - Cléa Rouillon
- Department of Gastroenterology, Caen University Hospital, Caen, France
| | - Francisca Joly
- Department of Gastroenterology, IBD unit, Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, Clichy, France
| | - Adrienne Elmorjani
- Genomic Medecine of Rare Diseases, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Julie Steffann
- Genomic Medecine of Rare Diseases, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Nadine Cerf-Bensussan
- Université Paris-Cité, Institut Imagine, Laboratory of Intestinal Immunity, INSERM U1163, Paris, France
| | - Marianna Parlato
- Université Paris-Cité, Institut Imagine, Laboratory of Intestinal Immunity, INSERM U1163, Paris, France.
| | - Fabienne Charbit-Henrion
- Université Paris-Cité, Institut Imagine, Laboratory of Intestinal Immunity, INSERM U1163, Paris, France.
- Genomic Medecine of Rare Diseases, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France.
| |
Collapse
|
41
|
Ott N, Faletti L, Heeg M, Andreani V, Grimbacher B. JAKs and STATs from a Clinical Perspective: Loss-of-Function Mutations, Gain-of-Function Mutations, and Their Multidimensional Consequences. J Clin Immunol 2023:10.1007/s10875-023-01483-x. [PMID: 37140667 DOI: 10.1007/s10875-023-01483-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 04/01/2023] [Indexed: 05/05/2023]
Abstract
The JAK/STAT signaling pathway plays a key role in cytokine signaling and is involved in development, immunity, and tumorigenesis for nearly any cell. At first glance, the JAK/STAT signaling pathway appears to be straightforward. However, on closer examination, the factors influencing the JAK/STAT signaling activity, such as cytokine diversity, receptor profile, overlapping JAK and STAT specificity among non-redundant functions of the JAK/STAT complexes, positive regulators (e.g., cooperating transcription factors), and negative regulators (e.g., SOCS, PIAS, PTP), demonstrate the complexity of the pathway's architecture, which can be quickly disturbed by mutations. The JAK/STAT signaling pathway has been, and still is, subject of basic research and offers an enormous potential for the development of new methods of personalized medicine and thus the translation of basic molecular research into clinical practice beyond the use of JAK inhibitors. Gain-of-function and loss-of-function mutations in the three immunologically particularly relevant signal transducers STAT1, STAT3, and STAT6 as well as JAK1 and JAK3 present themselves through individual phenotypic clinical pictures. The established, traditional paradigm of loss-of-function mutations leading to immunodeficiency and gain-of-function mutation leading to autoimmunity breaks down and a more differentiated picture of disease patterns evolve. This review is intended to provide an overview of these specific syndromes from a clinical perspective and to summarize current findings on pathomechanism, symptoms, immunological features, and therapeutic options of STAT1, STAT3, STAT6, JAK1, and JAK3 loss-of-function and gain-of-function diseases.
Collapse
Affiliation(s)
- Nils Ott
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Laura Faletti
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maximilian Heeg
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Division of Biological Sciences, Department of Molecular Biology, University of California, La Jolla, San Diego, CA, USA
| | - Virginia Andreani
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bodo Grimbacher
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Clinic of Rheumatology and Clinical Immunology, Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- DZIF - German Center for Infection Research, Satellite Center Freiburg, Freiburg, Germany
- CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- RESIST - Cluster of Excellence 2155 to Hanover Medical School, Satellite Center Freiburg, Freiburg, Germany
| |
Collapse
|
42
|
Bucciol G, Meyts I. Inherited and acquired errors of type I interferon immunity govern susceptibility to COVID-19 and multisystem inflammatory syndrome in children. J Allergy Clin Immunol 2023; 151:832-840. [PMID: 36841740 PMCID: PMC9951110 DOI: 10.1016/j.jaci.2023.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/02/2023] [Accepted: 02/01/2023] [Indexed: 02/27/2023]
Abstract
Since the beginning of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)/coronavirus disease 2019 (COVID-19) pandemic, global sequencing efforts have led in the field of inborn errors of immunity, and inspired particularly by previous research on life-threatening influenza, they have revealed that known and novel inborn errors affecting type I interferon immunity underlie critical COVID-19 in up to 5% of cases. In addition, neutralizing autoantibodies against type I interferons have been identified in up to 20% of patients with critical COVID-19 who are older than 80 years and 20% of fatal cases, with a higher prevalence in men and individuals older than 70 years. Also, inborn errors impairing regulation of type I interferon responses and RNA degradation have been found as causes of multisystem inflammatory syndrome in children, a life-threatening hyperinflammatory condition complicating otherwise mild initial SARS-CoV-2 infection in children and young adults. Better understanding of these immunologic mechanisms can aid in designing treatments for severe COVID-19, multisystem inflammatory syndrome in children, long COVID, and neuro-COVID.
Collapse
Affiliation(s)
- Giorgia Bucciol
- Laboratory of Inborn Errors of Immunity, Department of Microbiology, Immunology and Transplantation, Katholieke Universiteit Leuven, Leuven, Belgium; Childhood Immunology, Department of Pediatrics, Leuven University Hospitals, Leuven, Belgium
| | - Isabelle Meyts
- Laboratory of Inborn Errors of Immunity, Department of Microbiology, Immunology and Transplantation, Katholieke Universiteit Leuven, Leuven, Belgium; Childhood Immunology, Department of Pediatrics, Leuven University Hospitals, Leuven, Belgium.
| |
Collapse
|
43
|
Tong SY, Fan K, Zhou ZW, Liu LY, Zhang SQ, Fu Y, Wang GZ, Zhu Y, Yu YC. mvPPT: A Highly Efficient and Sensitive Pathogenicity Prediction Tool for Missense Variants. GENOMICS, PROTEOMICS & BIOINFORMATICS 2023; 21:414-426. [PMID: 35940520 PMCID: PMC10626173 DOI: 10.1016/j.gpb.2022.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 05/19/2022] [Accepted: 07/29/2022] [Indexed: 06/15/2023]
Abstract
Next-generation sequencing technologies both boost the discovery of variants in the human genome and exacerbate the challenges of pathogenic variant identification. In this study, we developed Pathogenicity Prediction Tool for missense variants (mvPPT), a highly sensitive and accurate missense variant classifier based on gradient boosting. mvPPT adopts high-confidence training sets with a wide spectrum of variant profiles, and extracts three categories of features, including scores from existing prediction tools, frequencies (allele frequencies, amino acid frequencies, and genotype frequencies), and genomic context. Compared with established predictors, mvPPT achieves superior performance in all test sets, regardless of data source. In addition, our study also provides guidance for training set and feature selection strategies, as well as reveals highly relevant features, which may further provide biological insights into variant pathogenicity. mvPPT is freely available at http://www.mvppt.club/.
Collapse
Affiliation(s)
- Shi-Yuan Tong
- Jing'an District Central Hospital of Shanghai, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Ke Fan
- Jing'an District Central Hospital of Shanghai, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Zai-Wei Zhou
- Shanghai Xunyin Biotechnology Co., Ltd., Shanghai 201802, China
| | - Lin-Yun Liu
- Jing'an District Central Hospital of Shanghai, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Shu-Qing Zhang
- Jing'an District Central Hospital of Shanghai, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Yinghui Fu
- Jing'an District Central Hospital of Shanghai, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Guang-Zhong Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ying Zhu
- Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| | - Yong-Chun Yu
- Jing'an District Central Hospital of Shanghai, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
44
|
Giardino G, Romano R, Lougaris V, Castagnoli R, Cillo F, Leonardi L, La Torre F, Soresina A, Federici S, Cancrini C, Pacillo L, Toriello E, Cinicola BL, Corrente S, Volpi S, Marseglia GL, Pignata C, Cardinale F. Immune tolerance breakdown in inborn errors of immunity: Paving the way to novel therapeutic approaches. Clin Immunol 2023; 251:109302. [PMID: 36967025 DOI: 10.1016/j.clim.2023.109302] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 03/06/2023] [Accepted: 03/22/2023] [Indexed: 05/12/2023]
Abstract
Up to 25% of the patients with inborn errors of immunity (IEI) also exhibit immunodysregulatory features. The association of immune dysregulation and immunodeficiency may be explained by different mechanisms. The understanding of mechanisms underlying immune dysregulation in IEI has paved the way for the development of targeted treatments. In this review article, we will summarize the mechanisms of immune tolerance breakdown and the targeted therapeutic approaches to immune dysregulation in IEI.
Collapse
Affiliation(s)
- Giuliana Giardino
- Pediatric Section, Department of Translational Medical Sciences, Federico II University, Naples, Italy.
| | - Roberta Romano
- Pediatric Section, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Vassilios Lougaris
- Department of Clinical and Experimental Sciences, Pediatrics Clinic and Institute for Molecular Medicine A. Nocivelli, University of Brescia and ASST-Spedali Civili di Brescia, Brescia, Italy
| | - Riccardo Castagnoli
- Department of Pediatrics, Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Francesca Cillo
- Pediatric Section, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Lucia Leonardi
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Francesco La Torre
- Department of Pediatrics, Giovanni XXIII Pediatric Hospital, University of Bari, Bari, Italy
| | - Annarosa Soresina
- Unit of Pediatric Immunology, Pediatrics Clinic, University of Brescia, ASST Spedali Civili Brescia, Brescia, Italy
| | - Silvia Federici
- Division of Rheumatology, IRCCS, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Caterina Cancrini
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Research Unit of Primary Immunodeficiencies, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Lucia Pacillo
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Research Unit of Primary Immunodeficiencies, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Elisabetta Toriello
- Pediatric Section, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Bianca Laura Cinicola
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | | | - Stefano Volpi
- Center for Autoinflammatory Diseases and Immunodeficiency, IRCCS Istituto Giannina Gaslini, Università degli Studi di Genova, Genoa, Italy
| | - Gian Luigi Marseglia
- Department of Pediatrics, Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Claudio Pignata
- Pediatric Section, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Fabio Cardinale
- Department of Pediatrics, Giovanni XXIII Pediatric Hospital, University of Bari, Bari, Italy
| |
Collapse
|
45
|
Doggett K, Keating N, Dehkhoda F, Bidgood GM, Meza Guzman LG, Leong E, Kueh A, Nicola NA, Kershaw NJ, Babon JJ, Alexander WS, Nicholson SE. The SOCS1 KIR and SH2 domain are both required for suppression of cytokine signaling in vivo. Cytokine 2023; 165:156167. [PMID: 36934508 DOI: 10.1016/j.cyto.2023.156167] [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: 12/21/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/19/2023]
Abstract
Suppressor Of Cytokine Signaling (SOCS) 1 is a critical negative regulator of cytokine signaling and required to protect against an excessive inflammatory response. Genetic deletion of Socs1 results in unrestrained cytokine signaling and neonatal lethality, characterised by an inflammatory immune infiltrate in multiple organs. Overexpression and structural studies have suggested that the SOCS1 kinase inhibitory region (KIR) and Src homology 2 (SH2) domain are important for interaction with and inhibition of the receptor-associated JAK1, JAK2 and TYK2 tyrosine kinases, which initiate downstream signaling. To investigate the role of the KIR and SH2 domain in SOCS1 function, we independently mutated key conserved residues in each domain and analysed the impact on cytokine signaling, and the in vivo impact on SOCS1 function. Mutation of the SOCS1-KIR or SH2 domain had no impact on the integrity of the SOCS box complex, however, mutation within the phosphotyrosine binding pocket of the SOCS1-SH2 domain specifically disrupted SOCS1 interaction with phosphorylated JAK1. In contrast, mutation of the KIR did not affect the interaction with JAK1, but did prevent SOCS1 inhibition of JAK1 autophosphorylation. In human and mouse cell lines, both mutants impacted the ability of SOCS1 to restrain cytokine signaling, and crucially, Socs1-R105A and Socs1-F59A mice displayed a neonatal lethality and excessive inflammatory phenotype similar to Socs1-null mice. This study defines a critical and non-redundant role for both the KIR and SH2 domain in endogenous SOCS1 function.
Collapse
Affiliation(s)
- Karen Doggett
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia.
| | - Narelle Keating
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Farhad Dehkhoda
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Grace M Bidgood
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Lizeth G Meza Guzman
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Evelyn Leong
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Andrew Kueh
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Nicos A Nicola
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Nadia J Kershaw
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Jeffrey J Babon
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Warren S Alexander
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Sandra E Nicholson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia.
| |
Collapse
|
46
|
The link between rheumatic disorders and inborn errors of immunity. EBioMedicine 2023; 90:104501. [PMID: 36870198 PMCID: PMC9996386 DOI: 10.1016/j.ebiom.2023.104501] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/11/2022] [Accepted: 02/10/2023] [Indexed: 03/06/2023] Open
Abstract
Inborn errors of immunity (IEIs) are immunological disorders characterized by variable susceptibility to infections, immune dysregulation and/or malignancies, as a consequence of damaging germline variants in single genes. Though initially identified among patients with unusual, severe or recurrent infections, non-infectious manifestations and especially immune dysregulation in the form of autoimmunity or autoinflammation can be the first or dominant phenotypic aspect of IEIs. An increasing number of IEIs causing autoimmunity or autoinflammation, including rheumatic disease have been reported over the last decade. Despite their rarity, identification of those disorders provided insight into the pathomechanisms of immune dysregulation, which may be relevant for understanding the pathogenesis of systemic rheumatic disorders. In this review, we present novel IEIs primarily causing autoimmunity or autoinflammation along with their pathogenic mechanisms. In addition, we explore the likely pathophysiological and clinical relevance of IEIs in systemic rheumatic disorders.
Collapse
|
47
|
Ren Y, Wang Y, Fang L, Ma M, Ge L, Su C, Xin L, He J, Yang J, Liu X. Deregulation of PRDM5 promotes cell proliferation by regulating JAK/STAT signaling pathway through SOCS1 in human lung adenocarcinoma. Cancer Med 2023; 12:4568-4578. [PMID: 36127737 PMCID: PMC9972168 DOI: 10.1002/cam4.5251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/19/2022] [Accepted: 09/02/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND PRDM5 is considered a tumor suppressor in several types of solid tumors and is involved in multiple cellular processes. However, target genes regulated by PRDM5 in lung cancer and its potential mechanism are poorly defined. METHODS Survival analysis was conducted using Kaplan-Meier estimates based on the online databases. RNA-sequencing and bioinformatics analysis were performed to identify the differentially expressed genes in PRDM5-overexpressed A549 cells. RESULTS We observed deregulated PRDM5 in several lung adenocarcinoma cell lines and its association with a poor prognosis. PRDM5 overexpression inhibited the proliferation of lung adenocarcinoma cells in vitro and suppressed tumor growth in a xenograft model. PRDM5 upregulated the promoter activity of SOCS1, which then inhibited the phosphorylation of JAK2 and STAT3. CONCLUSIONS Our study suggests that the low expression of PRDM5 promotes the proliferation of lung adenocarcinoma cells by downregulating SOCS1 and then upregulating the JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Yuanyuan Ren
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ye Wang
- Sinovac Biotech Co. Ltd, Beijing, China
| | | | - Mengchu Ma
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lin Ge
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Chao Su
- University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Lingbiao Xin
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jinyan He
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jie Yang
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xin Liu
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
48
|
Vergneault H, Picard C, Georgin-Lavialle S. Break down the barriers of auto-inflammation: How to deal with a monogenic auto-inflammatory disease and immuno-haematological features in 2022? Immunol Suppl 2023; 168:1-17. [PMID: 36151885 DOI: 10.1111/imm.13579] [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: 02/12/2022] [Accepted: 09/13/2022] [Indexed: 12/27/2022]
Abstract
In the past few years, the spectrum of monogenic systemic auto-inflammatory diseases (MSAID) has widely expanded beyond the typical recurrent fever. Immuno-haematological features, as cytopenias, hypogammaglobulinemia, hypereosinophilia, lymphoproliferation and immunodeficiency, have been described in association of several MSAID. The objective of this review was to describe these particular MSAID. MSAID must be suspected in front of immuno-haematological features associated with non-infectious recurrent fever, chronic systemic inflammation, inflammatory cutaneous manifestations, arthritis or inflammatory bowel disease. Genes and cellular mechanisms involved are various but some of them are of special interest. Defects in actine regulation pathway are notably associated with cytopenia and immune deficiency. Because of their frequency, ADA2 deficiency and Vacuoles, E1-Enzyme, X-linked, auto-inflammatory, Somatic (VEXAS) syndrome deserve to be noticed. ADA2 deficiency results in polyarteritis nodosa-like presentation with a wide panel of manifestations including cytopenia(s), lymphoproliferation and immune deficiency. Neutrophilic dermatosis or chondritis associated with macrocytic anaemia or myelodysplasia should lead to screen for VEXAS. Of note, most of MSAID are associated with inflammatory anaemia. We proposed here a clinical and pragmatic approach of MSAID associated with immuno-haematological features.
Collapse
Affiliation(s)
- Hélène Vergneault
- Internal Medicine Department, APHP, Tenon Hospital, National Reference Center for Autoinflammatory Diseases and Inflammatory Amyloidosis (CEREMAIA), Sorbonne University, Paris, France
| | - Capucine Picard
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, APHP, Université de Paris, Paris, France.,Laboratory of Lymphocyte Activation and Susceptibility to EBV, INSERM UMR1163, Imagine Institute, Necker Hospital for Sick Children, Université de Paris, Paris, France
| | - Sophie Georgin-Lavialle
- Internal Medicine Department, APHP, Tenon Hospital, National Reference Center for Autoinflammatory Diseases and Inflammatory Amyloidosis (CEREMAIA), Sorbonne University, Paris, France
| |
Collapse
|
49
|
Sobah ML, Scott AC, Laird M, Koole C, Liongue C, Ward AC. Socs3b regulates the development and function of innate immune cells in zebrafish. Front Immunol 2023; 14:1119727. [PMID: 36969252 PMCID: PMC10030509 DOI: 10.3389/fimmu.2023.1119727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/23/2023] [Indexed: 03/29/2023] Open
Abstract
Introduction Suppressor of cytokine signaling 3 (SOCS3) is a critical component of the negative feedback regulation that controls signaling by cytokines and other factors thereby ensuring that important processes such as hematopoiesis and inflammation occur at appropriate levels. Methods To gain further insights into SOCS3 function, the zebrafish socs3b gene was investigated through analysis of a knockout line generated using CRISPR/Cas9-mediated genome editing. Results Zebrafish socs3b knockout embryos displayed elevated numbers of neutrophils during primitive and definitive hematopoiesis but macrophage numbers were not altered. However, the absence of socs3b reduced neutrophil functionality but enhanced macrophage responses. Adult socs3b knockout zebrafish displayed reduced survival that correlated with an eye pathology involving extensive infiltration of neutrophils and macrophages along with immune cell dysregulation in other tissues. Discussion These findings identify a conserved role for Socs3b in the regulation of neutrophil production and macrophage activation.
Collapse
Affiliation(s)
| | - Aimee C. Scott
- Institue for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC, Australia
| | - Miranda Laird
- School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Cassandra Koole
- School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Clifford Liongue
- School of Medicine, Deakin University, Geelong, VIC, Australia
- Institue for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC, Australia
| | - Alister C. Ward
- School of Medicine, Deakin University, Geelong, VIC, Australia
- Institue for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC, Australia
- *Correspondence: Alister C. Ward,
| |
Collapse
|
50
|
Gutierrez MJ, Nino G, Sun D, Restrepo-Gualteros S, Sadreameli SC, Fiorino EK, Wu E, Vece T, Hagood JS, Maglione PJ, Kurland G, Koumbourlis A, Sullivan KE. The lung in inborn errors of immunity: From clinical disease patterns to molecular pathogenesis. J Allergy Clin Immunol 2022; 150:1314-1324. [PMID: 36244852 PMCID: PMC9826631 DOI: 10.1016/j.jaci.2022.08.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 11/06/2022]
Abstract
In addition to being a vital organ for gas exchange, the lung is a crucial immune organ continuously exposed to the external environment. Genetic defects that impair immune function, called inborn errors of immunity (IEI), often have lung disease as the initial and/or primary manifestation. Common types of lung disease seen in IEI include infectious complications and a diverse group of diffuse interstitial lung diseases. Although lung damage in IEI has been historically ascribed to recurrent infections, contributions from potentially targetable autoimmune and inflammatory pathways are now increasingly recognized. This article provides a practical guide to identifying the diverse pulmonary disease patterns in IEI based on lung imaging and respiratory manifestations, and integrates this clinical information with molecular mechanisms of disease and diagnostic assessments in IEI. We cover the entire IEI spectrum, including immunodeficiencies and immune dysregulation with monogenic autoimmunity and autoinflammation, as well as recently described IEI with pulmonary manifestations. Although the pulmonary manifestations of IEI are highly relevant for all age groups, special emphasis is placed on the pediatric population, because initial presentations often occur during childhood. We also highlight the pivotal role of genetic testing in the diagnosis of IEI involving the lungs and the critical need to develop multidisciplinary teams for the challenging evaluation of these rare but potentially life-threatening disorders.
Collapse
Affiliation(s)
- Maria J Gutierrez
- Division of Pediatric Allergy, Immunology and Rheumatology, Johns Hopkins University, Baltimore, Md.
| | - Gustavo Nino
- Division of Pediatric Pulmonary and Sleep Medicine, Children's National Hospital, Washington, DC; Department of Pediatrics, George Washington University School of Medicine, Washington, DC
| | - Di Sun
- Division of Pediatric Allergy and Immunology, The Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Sonia Restrepo-Gualteros
- Department of Pediatrics, School of Medicine, Universidad Nacional de Colombia, Bogotá, Colombia; Division of Pediatric Pulmonology, Fundacion Hospital La Misericordia, Bogotá, Colombia
| | - Sarah C Sadreameli
- Division of Pediatric Pulmonology and Sleep Medicine, Johns Hopkins University, Baltimore, Md
| | - Elizabeth K Fiorino
- Departments of Science Education and Pediatrics, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY
| | - Eveline Wu
- Division of Pediatric Allergy, Immunology and Rheumatology, University of North Carolina, Chapel Hill, NC
| | - Timothy Vece
- Division of Pediatric Pulmonology, University of North Carolina, Chapel Hill, NC
| | - James S Hagood
- Division of Pediatric Pulmonology, University of North Carolina, Chapel Hill, NC
| | - Paul J Maglione
- Division of Allergy and Immunology, Boston University, Boston, Mass
| | - Geoffrey Kurland
- Division of Pediatric Pulmonology and Sleep Medicine, University of Pittsburgh, Pittsburgh, Pa
| | - Anastassios Koumbourlis
- Division of Pediatric Pulmonary and Sleep Medicine, Children's National Hospital, Washington, DC; Department of Pediatrics, George Washington University School of Medicine, Washington, DC
| | - Kathleen E Sullivan
- Division of Pediatric Allergy and Immunology, The Children's Hospital of Philadelphia, Philadelphia, Pa
| |
Collapse
|