1
|
Mohamed Nashrudin KN, Zainal Abidin MA, Ng SE, Razali H, Jawin V, Zakaria AF, Mohd Ali R, Othman IS, Mohamed Unni MN, Hung CYR, Ismail IH. RASGRP1 Deficiency Associated with Diffuse Mesangial Sclerosis Infantile Nephrotic Syndrome and Epstein-Barr Virus-Induced Hodgkin's Lymphoma. PEDIATRIC ALLERGY, IMMUNOLOGY, AND PULMONOLOGY 2025. [PMID: 39752212 DOI: 10.1089/ped.2024.0062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Background: RAS guanyl-releasing protein 1 (RASGRP1) deficiency is characterized by immune dysregulation and Epstein-Barr virus (EBV)-related lymphoproliferation. Diffuse mesangial sclerosis is one of the infrequent causes of infantile nephrotic syndrome. Case Presentation: Here, we described a 7-year-old girl who was diagnosed with diffuse mesangial sclerosis at 5 months old and subsequently developed chronic bilateral neck swelling at the age of 3 years. Clinical assessment and investigations revealed a complex clinical picture, including recurrent cervical lymphadenopathy and recurrent infections. Further evaluation revealed immunological deficiencies, autoimmune lymphoproliferative syndrome-like illness, chronic EBV infection, and ultimately Hodgkin lymphoma. Genetic testing identified a RASGRP1 homozygous loss-of-function variant with both parents being carriers. Conclusion: This is the first reported case of RASGRP1 deficiency in Malaysia, and we highlight the challenges clinicians face when the disease manifests in varied presentations.
Collapse
Affiliation(s)
- Khairoon Nisa Mohamed Nashrudin
- Clinical Immunology Unit, Faculty of Medicine and Health Sciences, Department of Paediatrics, Universiti Putra Malaysia, Selangor, Malaysia
- Advanced Medical Research in Allergy and Clinical Immunology (AMRAC), Hospital Sultan Abdul Aziz Shah, Universiti Putra Malaysia, Selangor, Malaysia
| | - Mohd Azri Zainal Abidin
- Clinical Immunology Unit, Faculty of Medicine and Health Sciences, Department of Paediatrics, Universiti Putra Malaysia, Selangor, Malaysia
- Advanced Medical Research in Allergy and Clinical Immunology (AMRAC), Hospital Sultan Abdul Aziz Shah, Universiti Putra Malaysia, Selangor, Malaysia
| | - Shi Eng Ng
- Department of Paediatrics, Hospital Sultan Ismail, Johor, Malaysia
| | - Hadibiah Razali
- Paediatric Haemato-Oncology Unit, Hospital Sultan Ismail, Johor, Malaysia
| | - Vida Jawin
- Paediatric Haemato-Oncology Unit, Hospital Sultan Ismail, Johor, Malaysia
| | - Atiqah Farah Zakaria
- Faculty of Medicine and Health Sciences, Department of Otorhinolaryngology-Head and Neck Surgery, Universiti Putra Malaysia, Selangor, Malaysia
| | - Razana Mohd Ali
- Faculty of Medicine and Health Sciences, Department of Pathology, Universiti Putra Malaysia, Selangor, Malaysia
| | - Ida Shahnaz Othman
- Paediatric Haemato-Oncology Unit, Hospital Tunku Azizah, Kuala Lumpur, Malaysia
| | | | | | - Intan Hakimah Ismail
- Clinical Immunology Unit, Faculty of Medicine and Health Sciences, Department of Paediatrics, Universiti Putra Malaysia, Selangor, Malaysia
- Advanced Medical Research in Allergy and Clinical Immunology (AMRAC), Hospital Sultan Abdul Aziz Shah, Universiti Putra Malaysia, Selangor, Malaysia
| |
Collapse
|
2
|
Zhang X, Liu Y, Yang R, Guo Y, Yan M, Xiao Y, Dong Y, Zhang R, Qin Y, Bu Y, Zhang Y, Gao H. Phosphorylation of RasGRP1 by Shc3 prevents RasGRP1 degradation and contributes to Ras/c-Jun activation in hepatocellular carcinoma. Mol Cell Biochem 2024; 479:2307-2321. [PMID: 37646951 DOI: 10.1007/s11010-023-04839-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 08/14/2023] [Indexed: 09/01/2023]
Abstract
Ras guanine nucleotide-releasing protein 1 (RasGRP1), a Ras activator, is upregulated in hepatocellular carcinoma (HCC) and other kinds of cancer and is associated with the poor prognosis of patients. However, little is known about the underlying regulatory mechanisms of RasGRP1 in the context of cancer. Here, we report that RasGRP1 physically interacted with the adaptor protein Src homolog and collagen homolog 3 (Shc3). Moreover, RasGRP1 C-terminus domain (aa 607-797) bound to the central collagen-homology 1 (CH1) domain of Shc3. Subsequently, Shc3 enhanced the RasGRP1 tyrosine phosphorylation rate and stability by inhibiting its ubiquitination. Notably, the phosphorylation-mimicking mutants of RasGRP1, RasGRP1 Y704A, and Y748A, rescued the phosphorylation and ubiquitination levels of RasGRP1 in HCC cells. Further investigation showed that the RasGRP1 and Shc3 interaction induced activation of Ras and c-Jun, resulting in cell proliferation in vitro. Moreover, the regulation of Shc3/RasGRP1/Ras/c-Jun signal transduction was confirmed in vivo using the subcutaneous xenograft mouse model. Thus, we propose that continuous Shc3 overexpression may be a possible mechanism for maintaining RasGRP1 stability and that persistent activation of Ras/c-Jun signaling through the interaction of RasGRP1 and Shc3 is a key event increasing cell proliferation. Our findings suggest that the interaction of RasGRP1 and Shc3 plays an important role in HCC tumorigenesis and suggests the potential clinical usage of novel biomarkers and therapeutic targets in HCC.
Collapse
Affiliation(s)
- Xinran Zhang
- Department of Pharmacy, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Yun Liu
- Key Laboratory of Cancer Prevention and Therapy, Department of Pediatric Oncology, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - Rui Yang
- Department of Critical Care Medicine, Tianjin First Central Hospital, Tianjin Institute of Emergency Medicine, Tianjin, 300192, China
| | - Yuanyuan Guo
- Department of Pharmacy, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Meiling Yan
- Department of Pharmacy, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Ying Xiao
- Department of Pharmacy, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Yunzhuo Dong
- Department of Pharmacy, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Ruixia Zhang
- Department of Pharmacy, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Yinpeng Qin
- Department of Pharmacy, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Yishan Bu
- Department of Pharmacy, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Yi Zhang
- Department of Pharmacy, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Huier Gao
- Department of Pharmacy, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China.
| |
Collapse
|
3
|
Yang XT, Yang WL, Lau YL. NGS data analysis for molecular diagnosis of Inborn Errors of Immunity. Semin Immunol 2024; 74-75:101901. [PMID: 39509871 DOI: 10.1016/j.smim.2024.101901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 10/01/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024]
Abstract
Inborn errors of immunity (IEI) encompass a group of disorders with a strong genetic component. Prompt and accurate diagnosis of these disorders is essential for effective clinical management. Next-generation sequencing (NGS) has significantly enhanced the diagnostic process by offering a comprehensive and scalable approach for identifying genomic variations causal for these disorders. Nevertheless, the bioinformatics analysis of NGS data poses several challenges. In this review, we explore these challenges and share our insights on addressing them, aiming to improve the overall diagnostic yield.
Collapse
Affiliation(s)
- X T Yang
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - W L Yang
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Y L Lau
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
4
|
Fan S, Kang B, Li S, Li W, Chen C, Chen J, Deng L, Chen D, Zhou J. Exploring the multifaceted role of RASGRP1 in disease: immune, neural, metabolic, and oncogenic perspectives. Cell Cycle 2024; 23:722-746. [PMID: 38865342 PMCID: PMC11229727 DOI: 10.1080/15384101.2024.2366009] [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: 05/02/2023] [Accepted: 11/25/2023] [Indexed: 06/14/2024] Open
Abstract
RAS guanyl releasing protein 1 (RASGRP1) is a guanine nucleotide exchange factor (GEF) characterized by the presence of a RAS superfamily GEF domain. It functions as a diacylglycerol (DAG)-regulated nucleotide exchange factor, specifically activating RAS through the exchange of bound GDP for GTP. Activation of RAS by RASGRP1 has a wide range of downstream effects at the cellular level. Thus, it is not surprising that many diseases are associated with RASGRP1 disorders. Here, we present an overview of the structure and function of RASGRP1, its crucial role in the development, expression, and regulation of immune cells, and its involvement in various signaling pathways. This review comprehensively explores the relationship between RASGRP1 and various diseases, elucidates the underlying molecular mechanisms of RASGRP1 in each disease, and identifies potential therapeutic targets. This study provides novel insights into the role of RASGRP1 in insulin secretion and highlights its potential as a therapeutic target for diabetes. The limitations and challenges associated with studying RASGRP1 in disease are also discussed.
Collapse
Affiliation(s)
- Shangzhi Fan
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Bo Kang
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Shaoqian Li
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Weiyi Li
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Canyu Chen
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jixiang Chen
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Lijing Deng
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Danjun Chen
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jiecan Zhou
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
5
|
Mansour R, El-Orfali Y, Saidu A, Al-Kalamouni H, Mardirossian H, Hanna-Wakim R, Abboud M, Massaad MJ. A novel homozygous mutation in RASGRP1 that predisposes to immune dysregulation and immunodeficiency associated with uncontrolled Epstein-Barr virus-induced B cell proliferation. Clin Immunol 2023; 257:109813. [PMID: 37898412 DOI: 10.1016/j.clim.2023.109813] [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: 07/31/2023] [Revised: 10/05/2023] [Accepted: 10/21/2023] [Indexed: 10/30/2023]
Abstract
BACKGROUND RASGRP1-deficiency results in an immune dysregulation and immunodeficiency that manifest as autoimmunity, lymphoproliferation, lymphopenia, defective T cell function, and increased incidence of Epstein-Bar Virus infections and lymphomas. OBJECTIVE To investigate the mechanism of autoimmune hemolytic anemia and infections in a male patient of consanguineous parents from Lebanon. METHODS Genetic diagnosis was obtained using next generation and Sanger sequencing. Protein expression and phosphorylation were determined by immunoblotting. T and B cell development and function were studied by flow cytometry. Cytokine and immunoglobulin secretions were quantified by enzyme-linked immunosorbent assay. RESULTS The patient suffered from severe lymphopenia especially affecting the T cell compartment. Genetic analysis revealed a homozygous insertion of adenine at position 1396_1397 in RASGRP1 that abolished protein expression and downstream Ras signaling. T cells from the patient showed severe activation defects resulting in uncontrolled Epstein-Bar Virus-induced B cell proliferation. B cells from the patient were normal. CONCLUSION This report expands the spectrum of mutations in patients with RasGRP1 deficiency, and provides evidence for the important role RasGRP1 plays in the ability of T cells to control Epstein-Bar Virus-induced B cell proliferation. CLINICAL IMPLICATIONS Following diagnosis, the patient will be maintained on oral valganciclovir and monitored regularly for Epstein-Bar Virus infections to avoid the development of Epstein-Bar Virus- induced B cell lymphoma. He is also candidate for hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Rana Mansour
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Youmna El-Orfali
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Adam Saidu
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Habib Al-Kalamouni
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hagop Mardirossian
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Rima Hanna-Wakim
- Division of Pediatric Infectious Diseases, Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon; Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Miguel Abboud
- Division of Pediatric Hematology Oncology, Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon; Children's Cancer Center of Lebanon, American University of Beirut Medical Center, Beirut, Lebanon
| | - Michel J Massaad
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Division of Pediatric Infectious Diseases, Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon; Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon; Research Center of Excellence in Immunity and Infections, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
6
|
Pan-Hammarström Q, Casanova JL. Human genetic and immunological determinants of SARS-CoV-2 and Epstein-Barr virus diseases in childhood: Insightful contrasts. J Intern Med 2023; 294:127-144. [PMID: 36906905 DOI: 10.1111/joim.13628] [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] [Indexed: 03/13/2023]
Abstract
There is growing evidence to suggest that severe disease in children infected with common viruses that are typically benign in other children can result from inborn errors of immunity or their phenocopies. Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a cytolytic respiratory RNA virus, can lead to acute hypoxemic COVID-19 pneumonia in children with inborn errors of type I interferon (IFN) immunity or autoantibodies against IFNs. These patients do not appear to be prone to severe disease during infection with Epstein-Barr virus (EBV), a leukocyte-tropic DNA virus that can establish latency. By contrast, various forms of severe EBV disease, ranging from acute hemophagocytosis to chronic or long-term illnesses, such as agammaglobulinemia and lymphoma, can manifest in children with inborn errors disrupting specific molecular bridges involved in the control of EBV-infected B cells by cytotoxic T cells. The patients with these disorders do not seem to be prone to severe COVID-19 pneumonia. These experiments of nature reveal surprising levels of redundancy of two different arms of immunity, with type I IFN being essential for host defense against SARS-CoV-2 in respiratory epithelial cells, and certain surface molecules on cytotoxic T cells essential for host defense against EBV in B lymphocytes.
Collapse
Affiliation(s)
| | - Jean-Laurent Casanova
- St Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Howard Hughes Medical Institute, New York, New York, USA
- Laboratory of Human Genetics of Infectious Diseases, Inserm, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
- Department of Pediatrics, Necker Hospital for Sick Children, Paris, France
| |
Collapse
|
7
|
Deng M, Mao H. Inborn errors of immunity in mainland China: the past, present and future. BMJ Paediatr Open 2023; 7:e002002. [PMID: 37474202 PMCID: PMC10357751 DOI: 10.1136/bmjpo-2023-002002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/12/2023] [Indexed: 07/22/2023] Open
Abstract
Inborn errors of immunity (IEI), also known as primary immunodeficiency diseases, comprise a group of rare genetic disorders that affect the development or/and function of the immune system. These disorders predispose individuals to recurrent infections, autoimmunity, cancer and immune dysregulations. The field of IEI diagnosis and treatment in mainland China has made significant strides in recent years due to advances in genome sequencing, genetics, immunology and treatment strategies. However, the accessibility and affordability of diagnostic facilities and precision treatments remain variable among different regions. With the increasing government emphasis on rare disease prevention, diagnosis, and treatment, the field of IEI is expected to progress further in mainland China. Herein, we reviewed the development and current state of IEI in mainland China, highlighting the achievements made, as well as opportunities and challenges that lie ahead.
Collapse
Affiliation(s)
- Mengyue Deng
- Department of Immunology, Beijing Children's Hospital of Capital Medical University, National Center for Children's Health of China, Beijing, China
| | - Huawei Mao
- Department of Immunology, Beijing Children's Hospital of Capital Medical University, National Center for Children's Health of China, Beijing, China
- Ministry of Education Key Laboratory of Major Diseases in Children, Beijing Key Laboratory for Genetics of Birth Defects, Beijing, China
| |
Collapse
|
8
|
A Focused Review of Ras Guanine Nucleotide-Releasing Protein 1 in Immune Cells and Cancer. Int J Mol Sci 2023; 24:ijms24021652. [PMID: 36675167 PMCID: PMC9864139 DOI: 10.3390/ijms24021652] [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: 11/14/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Four Ras guanine nucleotide-releasing proteins (RasGRP1 through 4) belong to the family of guanine nucleotide exchange factors (GEFs). RasGRPs catalyze the release of GDP from small GTPases Ras and Rap and facilitate their transition from an inactive GDP-bound to an active GTP-bound state. Thus, they regulate critical cellular responses via many downstream GTPase effectors. Similar to other RasGRPs, the catalytic module of RasGRP1 is composed of the Ras exchange motif (REM) and Cdc25 domain, and the EF hands and C1 domain contribute to its cellular localization and regulation. RasGRP1 can be activated by a diacylglycerol (DAG)-mediated membrane recruitment and protein kinase C (PKC)-mediated phosphorylation. RasGRP1 acts downstream of the T cell receptor (TCR), B cell receptors (BCR), and pre-TCR, and plays an important role in the thymocyte maturation and function of peripheral T cells, B cells, NK cells, mast cells, and neutrophils. The dysregulation of RasGRP1 is known to contribute to numerous disorders that range from autoimmune and inflammatory diseases and schizophrenia to neoplasia. Given its position at the crossroad of cell development, inflammation, and cancer, RASGRP1 has garnered interest from numerous disciplines. In this review, we outline the structure, function, and regulation of RasGRP1 and focus on the existing knowledge of the role of RasGRP1 in leukemia and other cancers.
Collapse
|
9
|
Chan KW, Wong CY, Leung D, Yang X, Fok SFS, Mak PHS, Yao L, Ma W, Mao H, Zhao X, Liang W, Singh S, Barbouche MR, He JX, Jiang LP, Liew WK, Le MHT, Muktiarti D, Santos-Ocampo FJ, Djidjik R, Belaid B, Ismail IH, Abdul Latiff AH, Lee WS, Chen TX, Liu J, Jin R, Wang X, Chien YH, Yu HH, Raj D, Raj R, Vaughan J, Urban M, van den Berg S, Eley B, Lee ACW, Isa MS, Ang EY, Lee BW, Yeoh AEJ, Shek LP, Quynh Le NN, Nguyen VAT, Phan Nguyen Lien A, Capulong RD, Mallillin JM, Villanueva JCMM, Camonayan KAB, Vera MD, Casis-Hao RJ, Lobo RCM, Foronda R, Binas VWE, Boushaki S, Kechout N, Phongsamart G, Wongwaree S, Jiratchaya C, Lao-Araya M, Trakultivakorn M, Suratannon N, Jirapongsananuruk O, Chantveerawong T, Kamchaisatian W, Chan LL, Koh MT, Wong KJ, Fong SM, Thong MK, Latiff ZA, Noh LM, de Silva R, Jouhadi Z, Al-Saad K, Vignesh P, Jindal AK, Rawat A, Gupta A, Suri D, Yang J, Au EYL, Kwok JSY, Chan SY, Hui WYF, Chua GT, Duque JR, Cheong KN, Chong PCY, Ho MHK, Lee TL, Wong WHS, Yang W, Lee PP, Tu W, Yang XQ, Lau YL. Targeted Gene Sanger Sequencing Should Remain the First-Tier Genetic Test for Children Suspected to Have the Five Common X-Linked Inborn Errors of Immunity. Front Immunol 2022; 13:883446. [PMID: 35874699 PMCID: PMC9304939 DOI: 10.3389/fimmu.2022.883446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/02/2022] [Indexed: 11/13/2022] Open
Abstract
To address inborn errors of immunity (IEI) which were underdiagnosed in resource-limited regions, our centre developed and offered free genetic testing for the most common IEI by Sanger sequencing (SS) since 2001. With the establishment of The Asian Primary Immunodeficiency (APID) Network in 2009, the awareness and definitive diagnosis of IEI were further improved with collaboration among centres caring for IEI patients from East and Southeast Asia. We also started to use whole exome sequencing (WES) for undiagnosed cases and further extended our collaboration with centres from South Asia and Africa. With the increased use of Next Generation Sequencing (NGS), we have shifted our diagnostic practice from SS to WES. However, SS was still one of the key diagnostic tools for IEI for the past two decades. Our centre has performed 2,024 IEI SS genetic tests, with in-house protocol designed specifically for 84 genes, in 1,376 patients with 744 identified to have disease-causing mutations (54.1%). The high diagnostic rate after just one round of targeted gene SS for each of the 5 common IEI (X-linked agammaglobulinemia (XLA) 77.4%, Wiskott–Aldrich syndrome (WAS) 69.2%, X-linked chronic granulomatous disease (XCGD) 59.5%, X-linked severe combined immunodeficiency (XSCID) 51.1%, and X-linked hyper-IgM syndrome (HIGM1) 58.1%) demonstrated targeted gene SS should remain the first-tier genetic test for the 5 common X-linked IEI.
Collapse
Affiliation(s)
- Koon-Wing Chan
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Chung-Yin Wong
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Daniel Leung
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Xingtian Yang
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Susanna F. S. Fok
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Priscilla H. S. Mak
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Lei Yao
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Wen Ma
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Huawei Mao
- Department of Immunology, Ministry of Education Key Laboratory of Major Diseases in Children, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Xiaodong Zhao
- Children’s Hospital, Chongqing Medical University, Chongqing, China
| | - Weiling Liang
- Shenzhen Primary Immunodeficiency Diagnostic and Therapeutic Laboratory, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Surjit Singh
- Pediatric Allergy and Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | | - Jian-Xin He
- Department of Respiratory Medicine, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Li-Ping Jiang
- Children’s Hospital, Chongqing Medical University, Chongqing, China
| | - Woei-Kang Liew
- Department of Paediatric Medicine, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Minh Huong Thi Le
- Department of Immuno-Allergology and Rheumatology, National Hospital of Paediatrics, Hanoi, Vietnam
| | - Dina Muktiarti
- Department of Child Health, Faculty of Medicine Universitas Indonesia-Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | | | - Reda Djidjik
- Department of Medical Immunology, Beni Messous University Hospital Centre, University of Algiers 1, Algiers, Algeria
| | - Brahim Belaid
- Department of Medical Immunology, Beni Messous University Hospital Centre, University of Algiers 1, Algiers, Algeria
| | - Intan Hakimah Ismail
- Clinical Immunology Unit, Department of Paediatrics, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | | | - Way Seah Lee
- Department of Paediatrics, Faculty of Medicine, University Malaya, Kuala Lumpur, Malaysia
| | - Tong-Xin Chen
- Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jinrong Liu
- Department of Respiratory Medicine, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Runming Jin
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaochuan Wang
- Department of Clinical Immunology, Children’s Hospital of Fudan University, Shanghai, China
| | - Yin Hsiu Chien
- Department of Medical Genetics and Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsin-Hui Yu
- Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, National Taiwan University Children’s Hospital, Taipei, Taiwan
| | - Dinesh Raj
- Department of Paediatrics, Holy Family Hospital, University of Delhi, New Delhi, India
| | - Revathi Raj
- Department of Paediatric Haematology, Oncology, Blood and Marrow Transplantation, Apollo Hospitals, Chennai, India
| | - Jenifer Vaughan
- Department of Molecular Medicine and Haematology, National Health Laboratory Services, University of the Witwatersrand, Johannesburg, South Africa
| | - Michael Urban
- Division of Molecular Biology and Human Genetics, University of Stellenbosch Western Cape, Pretoria, South Africa
| | - Sylvia van den Berg
- Department of Immunology, Ampath and Department of Paediatrics and Child Health, University of Pretoria and Steve Biko Academic Hospital, Pretoria, South Africa
| | - Brian Eley
- Department of Paediatrics and Child Health, University of Cape Town and Red Cross War Memorial Children’s Hospital, Cape Town, South Africa
| | - Anselm Chi-Wai Lee
- Children’s Haematology and Cancer Center, Mount Elizabeth Hospital, Singapore, Singapore
| | - Mas Suhaila Isa
- Khoo Teck Puat-National University Children’s Medical Institute, National University Health System, Singapore, Singapore
| | - Elizabeth Y. Ang
- Khoo Teck Puat-National University Children’s Medical Institute, National University Health System, Singapore, Singapore
| | - Bee Wah Lee
- Khoo Teck Puat-National University Children’s Medical Institute, National University Health System, Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Allen Eng Juh Yeoh
- Khoo Teck Puat-National University Children’s Medical Institute, National University Health System, Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lynette P. Shek
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | | | - Van Anh Thi Nguyen
- Department of Rheumatology, Allergy, and Immunology, Vietnam National Children's Hospital, Hanoi, Vietnam
| | | | | | - Joanne Michelle Mallillin
- Child and Adult Allergy, Asthma and Immunology General Emilio Aguinaldo Memorial Hospital, Cavite, Philippines
| | - Jose Carlo Miguel M. Villanueva
- Section of Allergy and Clinical Immunology, Department of Pediatrics, University of Santo Tomas Hospital, Manila, Philippines
| | | | - Michelle De Vera
- Section of Allergy and Immunology, The Medical City, Pasig, Philippines
| | - Roxanne J. Casis-Hao
- Division of Allergy and Clinical Immunology, Department of Pediatrics, Philippine General Hospital, Manila, Philippines
| | - Rommel Crisenio M. Lobo
- Section of Allergy Asthma and Immunology, Fe del Mundo Medical Center, Quezon City, Philippines
| | - Ruby Foronda
- Department of Pediatrics, University of the East Ramon Magsaysay Memorial Medical Center, Quezon City, Philippines
| | | | - Soraya Boushaki
- Department of Medical Immunology, Beni Messous University Hospital Centre, University of Algiers 1, Algiers, Algeria
- Unit of Genetics, Laboratory of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Sciences and Technology “HouariBoumediene”, Algiers, Algeria
| | - Nadia Kechout
- Department of Immunology, Pasteur Institute of Algeria/Faculty of Medicine, Algiers, Algeria
| | - Gun Phongsamart
- Department of Pediatrics, Queen Sirikit National Institute of Child Health, Bangkok, Thailand
| | - Siriporn Wongwaree
- Department of Pediatrics, Queen Sirikit National Institute of Child Health, Bangkok, Thailand
| | - Chamnanrua Jiratchaya
- Department of Pediatrics, Queen Sirikit National Institute of Child Health, Bangkok, Thailand
| | - Mongkol Lao-Araya
- Department of Pediatrics, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Muthita Trakultivakorn
- Department of Pediatrics, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Narissara Suratannon
- Center of Excellence for Allergy and Clinical Immunology, Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Orathai Jirapongsananuruk
- Division of Allergy and Clinical Immunology, Department of Pediatrics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Teerapol Chantveerawong
- Division of Allergy and Clinical Immunology, Department of Medicine, Phramongkutklao Hospital, Bangkok, Thailand
| | - Wasu Kamchaisatian
- Division of Pediatrics Allergy and Immunology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Lee Lee Chan
- Subang Jaya Medical Centre, Subang Jaya, Malaysia
| | - Mia Tuang Koh
- Department of Paediatrics, Faculty of Medicine, University Malaya, Kuala Lumpur, Malaysia
| | - Ke Juin Wong
- Department of Paediatrics, Likas Hospital, Ministry of Health, Sabah, Malaysia
| | - Siew Moy Fong
- Department of Paediatrics, Likas Hospital, Ministry of Health, Sabah, Malaysia
| | - Meow-Keong Thong
- Genetics and Metabolism Unit, Department of Paediatrics, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Zarina Abdul Latiff
- Department of Pediatrics, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Lokman Mohd Noh
- Department of Pediatrics, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- Department of Paediatrics, Hospital Tunku Azizah, Ministry of Health Malaysia, Kuala Lumpur, Malaysia
| | - Rajiva de Silva
- Department of Immunology, Medical Research Institute, Colombo, Sri Lanka
| | - Zineb Jouhadi
- Department of Pediatric Infectious Diseases, Children’s Hospital CHU Ibn Rochd, University Hassan 2, Casablanca, Morocco
| | - Khulood Al-Saad
- Department of Pediatrics, Salmaniya Medical Complex, Manama, Bahrain
| | - Pandiarajan Vignesh
- Pediatric Allergy and Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ankur Kumar Jindal
- Pediatric Allergy and Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Amit Rawat
- Pediatric Allergy and Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Anju Gupta
- Pediatric Allergy and Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Deepti Suri
- Pediatric Allergy and Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Jing Yang
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Elaine Yuen-Ling Au
- Division of Clinical Immunology, Department of Pathology, Queen Mary Hospital, Hong Kong, Hong Kong SAR, China
| | - Janette Siu-Yin Kwok
- Division of Transplantation and Immunogenetics, Department of Pathology, Queen Mary Hospital, Hong Kong, Hong Kong SAR, China
| | - Siu-Yuen Chan
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Wayland Yuk-Fun Hui
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Gilbert T. Chua
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jaime Rosa Duque
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Kai-Ning Cheong
- Hong Kong Children’s Hospital, Hong Kong, Hong Kong SAR, China
| | | | | | - Tsz-Leung Lee
- Hong Kong Children’s Hospital, Hong Kong, Hong Kong SAR, China
| | - Wilfred Hing-Sang Wong
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Wanling Yang
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Pamela P. Lee
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Wenwei Tu
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Xi-Qiang Yang
- Children’s Hospital, Chongqing Medical University, Chongqing, China
| | - Yu Lung Lau
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- *Correspondence: Yu Lung Lau,
| |
Collapse
|
10
|
Zhang L, Chen Z, Li W, Liu Q, Wang Y, Chen X, Tian Z, Yang Q, An Y, Zhang Z, Mao H, Tang X, Lv G, Zhao X. Combined Immunodeficiency Caused by a Novel De Novo Gain-of-Function RAC2 Mutation. J Clin Immunol 2022; 42:1280-1292. [PMID: 35596857 DOI: 10.1007/s10875-022-01288-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 05/02/2022] [Indexed: 01/09/2023]
Abstract
Ras-related C3 botulinum toxin substrate 2 (RAC2) is a GTPase exclusively expressed in hematopoietic cells that acts as a pivotal regulator of several aspects of cell behavior via various cellular processes. RAC2 undergoes a tightly regulated GTP-binding/GTP-hydrolysis cycle, enabling it to function as a molecular switch. Mutations in RAC2 have been identified in 18 patients with different forms of primary immunodeficiency, ranging from phagocyte defects caused by dominant negative mutations to common variable immunodeficiency resulting from autosomal recessive loss-of-function mutations, or severe combined immunodeficiency due to dominant activating gain-of-function mutations. Here, we describe an 11-year-old girl with combined immunodeficiency presenting with recurrent respiratory infections and bronchiectasis. Immunological investigations revealed low T-cell receptor excision circle/K-deleting recombination excision circles numbers, lymphopenia, and low serum immunoglobulin G. Targeted next-generation sequencing identified a novel heterozygous mutation in RAC2, c.86C > G (p.P29R), located in the highly conserved Switch I domain. The mutation resulted in enhanced reactive oxygen species production, elevated F-actin content, and increased RAC2 protein expression in neutrophils, as well as increased cytokine production and a dysregulated phenotype in T lymphocytes. Furthermore, the dominant activating RAC2 mutation led to accelerated apoptosis with augmented intracellular active caspase 3, impaired actin polarization in lymphocytes and neutrophils, and diminished RAC2 polarization in neutrophils. We present a novel RAC2 gain-of-function mutation with implications for immunodeficiency and linked to functional dysregulation, including abnormal apoptosis and cell polarization arising from altered RAC2 expression. Thus, our findings broaden the spectrum of known RAC2 mutations and their underlying mechanisms.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing); China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China.,Division of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China.,Department of Nephrology, Rheumatology and Immunology, Hunan Children's Hospital, The Paediatric Academy of University of South China, Changsha, Hunan, China
| | - Zhi Chen
- Department of Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing); China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China.,Division of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China.,Department of Immunology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wenyan Li
- Department of Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing); China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China.,Division of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Qiao Liu
- Department of Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing); China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China.,Division of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yanping Wang
- Department of Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing); China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China.,Division of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xuemei Chen
- Department of Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing); China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China.,Division of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Zhirui Tian
- Department of Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing); China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China.,Division of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Qiuyun Yang
- Department of Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing); China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China.,Division of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yunfei An
- Department of Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing); China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China.,Division of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Zhiyong Zhang
- Department of Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing); China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China.,Division of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Huawei Mao
- Department of Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing); China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China.,Division of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xuemei Tang
- Department of Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing); China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China.,Division of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Ge Lv
- Department of Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing); China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China. .,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China. .,Division of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China.
| | - Xiaodong Zhao
- Department of Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing); China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China. .,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China. .,Division of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
11
|
Two Novel Biallelic RASGRP1 Mutations Presenting with Immunodeficiency, Hodgkin's Lymphoma, and Autoimmunity. J Clin Immunol 2022; 42:1160-1163. [PMID: 35593944 DOI: 10.1007/s10875-022-01281-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 04/26/2022] [Indexed: 10/18/2022]
|
12
|
Kuskonmaz B, Ayvaz D, Okur FV, Aydın B, Tezcan I, Uckan Cetinkaya D. First allogeneic hematopoietic stem cell transplantation in RASGRP1 deficiency: long-term follow-up. Bone Marrow Transplant 2022; 57:1210-1212. [PMID: 35568755 PMCID: PMC9106979 DOI: 10.1038/s41409-022-01704-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 04/24/2022] [Accepted: 05/03/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Baris Kuskonmaz
- Faculty of Medicine, Department of Pediatrics, Division of BMT Unit, Hacettepe University, Ankara, Turkey.
| | - Deniz Ayvaz
- Faculty of Medicine, Department of Pediatrics, Division of Immunology, Hacettepe University, Ankara, Turkey
| | - Fatma Visal Okur
- Faculty of Medicine, Department of Pediatrics, Division of BMT Unit, Hacettepe University, Ankara, Turkey
| | - Burça Aydın
- Faculty of Medicine, Department of Pediatrics, Division of Oncology, Hacettepe University, Ankara, Turkey
| | - Ilhan Tezcan
- Faculty of Medicine, Department of Pediatrics, Division of Immunology, Hacettepe University, Ankara, Turkey
| | - Duygu Uckan Cetinkaya
- Faculty of Medicine, Department of Pediatrics, Division of BMT Unit, Hacettepe University, Ankara, Turkey
| |
Collapse
|
13
|
Li Y, Deng M, Li Y, Mao X, Yan S, Tang X, Mao H. Clinical heterogeneity of NLRP12-associated autoinflammatory diseases. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
14
|
Latour S. Inherited immunodeficiencies associated with proximal and distal defects in T cell receptor signaling and co-signaling. Biomed J 2022; 45:321-333. [PMID: 35091087 PMCID: PMC9250091 DOI: 10.1016/j.bj.2022.01.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 12/24/2022] Open
Affiliation(s)
- Sylvain Latour
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, Inserm UMR 1163, Université de Paris, Institut Imagine, Paris, France.
| |
Collapse
|
15
|
Redmond MT, Scherzer R, Prince BT. Novel Genetic Discoveries in Primary Immunodeficiency Disorders. Clin Rev Allergy Immunol 2022; 63:55-74. [PMID: 35020168 PMCID: PMC8753955 DOI: 10.1007/s12016-021-08881-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2021] [Indexed: 01/12/2023]
Abstract
The field of Immunology is one that has undergone great expansion in recent years. With the advent of new diagnostic modalities including a variety of genetic tests (discussed elsewhere in this journal), the ability to diagnose a patient with a primary immunodeficiency disorder (PIDD) has become a more streamlined process. With increased availability of genetic testing for those with suspected or known PIDD, there has been a significant increase in the number of genes associated with this group of disorders. This is of great importance as a misdiagnosis of these rare diseases can lead to a delay in what can be critical treatment options. At times, those options can include life-saving medications or procedures. Presentation of patients with PIDD can vary greatly based on the specific genetic defect and the part(s) of the immune system that is affected by the variation. PIDD disorders lead to varying levels of increased risk of infection ranging from a mild increase such as with selective IgA deficiency to a profound risk with severe combined immunodeficiency. These diseases can also cause a variety of other clinical findings including autoimmunity and gastrointestinal disease.
Collapse
Affiliation(s)
- Margaret T. Redmond
- Division of Allergy and Immunology, Department of Pediatrics, Nationwide Children’s Hospital, The Ohio State University College of Medicine, Columbus, OH USA
| | - Rebecca Scherzer
- Division of Allergy and Immunology, Department of Pediatrics, Nationwide Children’s Hospital, The Ohio State University College of Medicine, Columbus, OH USA
| | - Benjamin T. Prince
- Division of Allergy and Immunology, Department of Pediatrics, Nationwide Children’s Hospital, The Ohio State University College of Medicine, Columbus, OH USA
| |
Collapse
|
16
|
Lino CNR, Ghosh S. Epstein-Barr Virus in Inborn Immunodeficiency-More Than Infection. Cancers (Basel) 2021; 13:cancers13194752. [PMID: 34638238 PMCID: PMC8507541 DOI: 10.3390/cancers13194752] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/05/2021] [Accepted: 09/06/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Epstein–Barr Virus (EBV) is a common virus that is readily controlled by a healthy immune system and rarely causes serious problems in infected people. However, patients with certain genetic defects of their immune system might have difficulties controlling EBV and often develop severe and life-threatening conditions, such as severe inflammation and malignancies. In this review, we provide a summary of inherited immune diseases that lead to a high susceptibility to EBV infection and discuss how this infection is associated with cancer development. Abstract Epstein–Barr Virus (EBV) is a ubiquitous virus affecting more than 90% of the world’s population. Upon infection, it establishes latency in B cells. It is a rather benign virus for immune-competent individuals, in whom infections usually go unnoticed. Nevertheless, EBV has been extensively associated with tumorigenesis. Patients suffering from certain inborn errors of immunity are at high risk of developing malignancies, while infection in the majority of immune-competent individuals does not seem to lead to immune dysregulation. Herein, we discuss how inborn mutations in TNFRSF9, CD27, CD70, CORO1A, CTPS1, ITK, MAGT1, RASGRP1, STK4, CARMIL2, SH2D1A, and XIAP affect the development, differentiation, and function of key factors involved in the immunity against EBV, leading to increased susceptibility to lymphoproliferative disease and lymphoma.
Collapse
Affiliation(s)
| | - Sujal Ghosh
- Correspondence: ; Tel.: +49-211-811-6224; Fax: +49-211-811-6191
| |
Collapse
|
17
|
López-Nevado M, González-Granado LI, Ruiz-García R, Pleguezuelo D, Cabrera-Marante O, Salmón N, Blanco-Lobo P, Domínguez-Pinilla N, Rodríguez-Pena R, Sebastián E, Cruz-Rojo J, Olbrich P, Ruiz-Contreras J, Paz-Artal E, Neth O, Allende LM. Primary Immune Regulatory Disorders With an Autoimmune Lymphoproliferative Syndrome-Like Phenotype: Immunologic Evaluation, Early Diagnosis and Management. Front Immunol 2021; 12:671755. [PMID: 34447369 PMCID: PMC8382720 DOI: 10.3389/fimmu.2021.671755] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 07/16/2021] [Indexed: 12/26/2022] Open
Abstract
Primary immune regulatory disorders (PIRD) are associated with autoimmunity, autoinflammation and/or dysregulation of lymphocyte homeostasis. Autoimmune lymphoproliferative syndrome (ALPS) is a PIRD due to an apoptotic defect in Fas-FasL pathway and characterized by benign and chronic lymphoproliferation, autoimmunity and increased risk of lymphoma. Clinical manifestations and typical laboratory biomarkers of ALPS have also been found in patients with a gene defect out of the Fas-FasL pathway (ALPS-like disorders). Following the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA), we identified more than 600 patients suffering from 24 distinct genetic defects described in the literature with an autoimmune lymphoproliferative phenotype (ALPS-like syndromes) corresponding to phenocopies of primary immunodeficiency (PID) (NRAS, KRAS), susceptibility to EBV (MAGT1, PRKCD, XIAP, SH2D1A, RASGRP1, TNFRSF9), antibody deficiency (PIK3CD gain of function (GOF), PIK3R1 loss of function (LOF), CARD11 GOF), regulatory T-cells defects (CTLA4, LRBA, STAT3 GOF, IL2RA, IL2RB, DEF6), combined immunodeficiencies (ITK, STK4), defects in intrinsic and innate immunity and predisposition to infection (STAT1 GOF, IL12RB1) and autoimmunity/autoinflammation (ADA2, TNFAIP3,TPP2, TET2). CTLA4 and LRBA patients correspond around to 50% of total ALPS-like cases. However, only 100% of CTLA4, PRKCD, TET2 and NRAS/KRAS reported patients had an ALPS-like presentation, while the autoimmunity and lymphoproliferation combination resulted rare in other genetic defects. Recurrent infections, skin lesions, enteropathy and malignancy are the most common clinical manifestations. Some approaches available for the immunological study and identification of ALPS-like patients through flow cytometry and ALPS biomarkers are provided in this work. Protein expression assays for NKG2D, XIAP, SAP, CTLA4 and LRBA deficiencies and functional studies of AKT, STAT1 and STAT3 phosphorylation, are showed as useful tests. Patients suspected to suffer from one of these disorders require rapid and correct diagnosis allowing initiation of tailored specific therapeutic strategies and monitoring thereby improving the prognosis and their quality of life.
Collapse
Affiliation(s)
- Marta López-Nevado
- Immunology Department, University Hospital 12 de Octubre, Madrid, Spain
- Research Institute Hospital 12 Octubre (imas12), Madrid, Spain
| | - Luis I. González-Granado
- Research Institute Hospital 12 Octubre (imas12), Madrid, Spain
- Immunodeficiency Unit, Department of Pediatrics, University Hospital 12 de Octubre, Madrid, Spain
| | - Raquel Ruiz-García
- Immunology Department, Centre Diagnòstic Biomèdic, Hospital Clínic, Barcelona, Spain
| | - Daniel Pleguezuelo
- Immunology Department, University Hospital 12 de Octubre, Madrid, Spain
- Research Institute Hospital 12 Octubre (imas12), Madrid, Spain
| | - Oscar Cabrera-Marante
- Immunology Department, University Hospital 12 de Octubre, Madrid, Spain
- Research Institute Hospital 12 Octubre (imas12), Madrid, Spain
| | - Nerea Salmón
- Research Institute Hospital 12 Octubre (imas12), Madrid, Spain
- Immunodeficiency Unit, Department of Pediatrics, University Hospital 12 de Octubre, Madrid, Spain
| | - Pilar Blanco-Lobo
- Paediatric Infectious Diseases, Rheumatology and Immunology Unit, University Hospital Virgen del Rocío, Institute of Biomedicine, Biomedicine Institute (IBiS)/University of Seville/Superior Council of Scientific Investigations (CSIC), Seville, Spain
| | - Nerea Domínguez-Pinilla
- Research Institute Hospital 12 Octubre (imas12), Madrid, Spain
- Pediatric Hematology and Oncology Unit, Toledo Hospital Complex, Toledo, Spain and University Hospital 12 de Octubre, Madrid, Spain
| | | | - Elena Sebastián
- Hematology and Hemotherapy Unit, University Children’s Hospital Niño Jesús, Madrid, Spain
| | - Jaime Cruz-Rojo
- Endocrine Unit, Department of Pediatrics, University Hospital 12 de Octubre, Madrid, Spain
| | - Peter Olbrich
- Paediatric Infectious Diseases, Rheumatology and Immunology Unit, University Hospital Virgen del Rocío, Institute of Biomedicine, Biomedicine Institute (IBiS)/University of Seville/Superior Council of Scientific Investigations (CSIC), Seville, Spain
| | - Jesús Ruiz-Contreras
- Research Institute Hospital 12 Octubre (imas12), Madrid, Spain
- Immunodeficiency Unit, Department of Pediatrics, University Hospital 12 de Octubre, Madrid, Spain
- School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Estela Paz-Artal
- Immunology Department, University Hospital 12 de Octubre, Madrid, Spain
- Research Institute Hospital 12 Octubre (imas12), Madrid, Spain
- School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Olaf Neth
- Paediatric Infectious Diseases, Rheumatology and Immunology Unit, University Hospital Virgen del Rocío, Institute of Biomedicine, Biomedicine Institute (IBiS)/University of Seville/Superior Council of Scientific Investigations (CSIC), Seville, Spain
| | - Luis M. Allende
- Immunology Department, University Hospital 12 de Octubre, Madrid, Spain
- Research Institute Hospital 12 Octubre (imas12), Madrid, Spain
- School of Medicine, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
18
|
Human inborn errors of immunity to oncogenic viruses. Curr Opin Immunol 2021; 72:277-285. [PMID: 34364035 DOI: 10.1016/j.coi.2021.06.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/21/2021] [Accepted: 06/30/2021] [Indexed: 12/25/2022]
Abstract
Oncoviruses are viruses that can cause tumors. Seven viruses are currently recognized as oncogenic in humans: Epstein Barr virus (EBV), Kaposi sarcoma-associated herpesvirus (KSHV, also known as HHV8), human papillomaviruses (HPVs), hepatitis B virus (HBV), hepatitis C virus (HCV), human T-lymphotropic virus-1 (HTLV-1), and Merkel cell polyomavirus (MCPyV). The clinical phenotypes resulting from infection with these oncoviruses range from asymptomatic infection to invasive cancers. Patients with inborn errors of immunity (IEI) are prone to the development of infectious diseases caused by a narrow or broad spectrum of pathogens, including oncoviruses in some cases. Studies of patients with IEI have deepened our understanding of the non-redundant mechanisms underlying the control of EBV, HHV8 and HPV infections. The human genetic factors conferring predisposition to oncogenic HBV, HCV, HTLV-1 and MCPyV manifestations remain elusive. We briefly review here what is currently known about the IEI conferring predisposition to severe infection with oncoviruses.
Collapse
|
19
|
Dieudonne Y, Martin M, Korganow AS, Boutboul D, Guffroy A. [EBV and immunodeficiency]. Rev Med Interne 2021; 42:832-843. [PMID: 33867195 DOI: 10.1016/j.revmed.2021.03.324] [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: 12/05/2020] [Revised: 03/18/2021] [Accepted: 03/21/2021] [Indexed: 11/30/2022]
Abstract
Epstein-Barr virus (EBV), discovered in 1964, is a double-stranded DNA virus belonging to the Herpesviridae family. EBV has a lymphoid tropism with transforming capacities using different oncogenic viral proteins. This virus has two replication cycles: a lytic cycle mainly occuring during primary infection and a latent cycle allowing viral persistence into host memory B cells. More than 90% of adults are seropositive for EBV worldwide, with a past history of asymptomatic or mild primary infection. EBV infection can sometimes cause life-threatening complications such as hemophagocytic lymphohistiocytosis, and lead to the development of lymphoproliferative disorders or cancers. Risk factors associated with these phenotypes have been recently described through the study of monogenic primary immune deficiencies with EBV susceptibility. We here review the virological and immunological aspects of EBV infection and EBV-related complications with an overview of current available treatments.
Collapse
Affiliation(s)
- Y Dieudonne
- Université de Strasbourg, Inserm UMR - S1109, 67000 Strasbourg, France; Hôpitaux universitaires de Strasbourg, service d'immunologie clinique et de médecine interne, centre national de référence des maladies auto-immunes et systémiques rares, Est/Sud-Ouest (RESO), centre de compétence pour les déficits immunitaires primitifs de l'adulte, 67000 Strasbourg, France; Université de Strasbourg, faculté de médecine, 67000 Strasbourg, France
| | - M Martin
- Service de médecine interne, maladies infectieuses et tropicales, centre hospitalier universitaire de Poitiers, 86021 Poitiers, France; Université de Poitiers, 86021 Poitiers, France
| | - A-S Korganow
- Université de Strasbourg, Inserm UMR - S1109, 67000 Strasbourg, France; Hôpitaux universitaires de Strasbourg, service d'immunologie clinique et de médecine interne, centre national de référence des maladies auto-immunes et systémiques rares, Est/Sud-Ouest (RESO), centre de compétence pour les déficits immunitaires primitifs de l'adulte, 67000 Strasbourg, France; Université de Strasbourg, faculté de médecine, 67000 Strasbourg, France
| | - D Boutboul
- Service d'immunopathologie clinique, U976 HIPI, hôpital Saint-Louis, université de Paris, Paris, France.
| | - A Guffroy
- Université de Strasbourg, Inserm UMR - S1109, 67000 Strasbourg, France; Hôpitaux universitaires de Strasbourg, service d'immunologie clinique et de médecine interne, centre national de référence des maladies auto-immunes et systémiques rares, Est/Sud-Ouest (RESO), centre de compétence pour les déficits immunitaires primitifs de l'adulte, 67000 Strasbourg, France; Université de Strasbourg, faculté de médecine, 67000 Strasbourg, France.
| |
Collapse
|
20
|
Baars MJ, Douma T, Simeonov DR, Myers DR, Kulhanek K, Banerjee S, Zwakenberg S, Baltissen MP, Amini M, de Roock S, van Wijk F, Vermeulen M, Marson A, Roose JP, Vercoulen Y. Dysregulated RASGRP1 expression through RUNX1 mediated transcription promotes autoimmunity. Eur J Immunol 2021; 51:471-482. [PMID: 33065764 PMCID: PMC7894479 DOI: 10.1002/eji.201948451] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 08/11/2020] [Accepted: 10/15/2020] [Indexed: 12/19/2022]
Abstract
RasGRP1 is a Ras guanine nucleotide exchange factor, and an essential regulator of lymphocyte receptor signaling. In mice, Rasgrp1 deletion results in defective T lymphocyte development. RASGRP1-deficient patients suffer from immune deficiency, and the RASGRP1 gene has been linked to autoimmunity. However, how RasGRP1 levels are regulated, and if RasGRP1 dosage alterations contribute to autoimmunity remains unknown. We demonstrate that diminished Rasgrp1 expression caused defective T lymphocyte selection in C57BL/6 mice, and that the severity of inflammatory disease inversely correlates with Rasgrp1 expression levels. In patients with autoimmunity, active inflammation correlated with decreased RASGRP1 levels in CD4+ T cells. By analyzing H3K27 acetylation profiles in human T cells, we identified a RASGRP1 enhancer that harbors autoimmunity-associated SNPs. CRISPR-Cas9 disruption of this enhancer caused lower RasGRP1 expression, and decreased binding of RUNX1 and CBFB transcription factors. Analyzing patients with autoimmunity, we detected reduced RUNX1 expression in CD4+ T cells. Lastly, we mechanistically link RUNX1 to transcriptional regulation of RASGRP1 to reveal a key circuit regulating RasGRP1 expression, which is vital to prevent inflammatory disease.
Collapse
Affiliation(s)
- Matthijs J.D. Baars
- Molecular Cancer Research, Center for Molecular MedicineUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Thera Douma
- Center of Translational ImmunologyUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Dimitre R. Simeonov
- Diabetes CenterUniversity of California San FranciscoSan FranciscoCAUSA
- Biomedical Sciences Graduate ProgramUniversity of California San FranciscoSan FranciscoCAUSA
| | - Darienne R. Myers
- Biomedical Sciences Graduate ProgramUniversity of California San FranciscoSan FranciscoCAUSA
- Department of AnatomyUniversity of California San FranciscoSan FranciscoCAUSA
| | - Kayla Kulhanek
- Department of AnatomyUniversity of California San FranciscoSan FranciscoCAUSA
| | - Saikat Banerjee
- Department of AnatomyUniversity of California San FranciscoSan FranciscoCAUSA
| | - Susan Zwakenberg
- Molecular Cancer Research, Center for Molecular MedicineUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Marijke P. Baltissen
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode InstituteRadboud University NijmegenNijmegenThe Netherlands
| | - Mojtaba Amini
- Molecular Cancer Research, Center for Molecular MedicineUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Sytze de Roock
- Pediatric Immunology and Rheumatology, Wilhelmina Children's HospitalUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Femke van Wijk
- Center of Translational ImmunologyUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
- Pediatric Immunology and Rheumatology, Wilhelmina Children's HospitalUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Michiel Vermeulen
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode InstituteRadboud University NijmegenNijmegenThe Netherlands
| | - Alexander Marson
- Diabetes CenterUniversity of California San FranciscoSan FranciscoCAUSA
- J. David Gladstone InstitutesSan FranciscoCAUSA
- Department of MedicineUniversity of CaliforniaSan FranciscoCAUSA
- Department of Microbiology and ImmunologyUniversity of CaliforniaSan FranciscoCAUSA
| | - Jeroen P. Roose
- Department of AnatomyUniversity of California San FranciscoSan FranciscoCAUSA
| | - Yvonne Vercoulen
- Molecular Cancer Research, Center for Molecular MedicineUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
21
|
Mohtashami M, Razavi A, Abolhassani H, Aghamohammadi A, Yazdani R. Primary Immunodeficiency and Thrombocytopenia. Int Rev Immunol 2021; 41:135-159. [PMID: 33464134 DOI: 10.1080/08830185.2020.1868454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Primary immunodeficiency (PID) or Inborn errors of immunity (IEI) refers to a heterogeneous group of disorders characterized by immune system impairment. Although patients with IEI manifest highly variable symptoms, the most common clinical manifestations are recurrent infections, autoimmunity and malignancies. Some patients present hematological abnormality including thrombocytopenia due to different pathogenic mechanisms. This review focuses on primary and secondary thrombocytopenia as a complication, which can occur in IEI. Based on the International Union of Immunological Societies phenotypic classification for IEI, the several innate and adaptive immunodeficiency disorders can lead to thrombocytopenia. This review, for the first time, describes manifestation, mechanism and therapeutic modalities for thrombocytopenia in different classes of IEI.
Collapse
Affiliation(s)
- Maryam Mohtashami
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran.,Research Center for Immunodeficiencies, Tehran University of Medical Sciences, Tehran, Iran
| | - Azadehsadat Razavi
- Research Center for Immunodeficiencies, Tehran University of Medical Sciences, Tehran, Iran.,Department of Animal Biology, Faculty of Biology Sciences, University of Kharazmi, Tehran, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hassan Abolhassani
- Research Center for Immunodeficiencies, Tehran University of Medical Sciences, Tehran, Iran.,Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Asghar Aghamohammadi
- Research Center for Immunodeficiencies, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Yazdani
- Research Center for Immunodeficiencies, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Sharifinejad N, Azizi G, Behniafard N, Zaki-Dizaji M, Jamee M, Yazdani R, Abolhassani H, Aghamohammadi A. Protein Kinase C-Delta Defect in Autoimmune Lymphoproliferative Syndrome-Like Disease: First Case from the National Iranian Registry and Review of the Literature. Immunol Invest 2020; 51:331-342. [PMID: 33047643 DOI: 10.1080/08820139.2020.1829638] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND Protein kinase C is a family of serine/threonine kinases that play a key role in the adaptive immune cell signaling, as well as regulation of growth, apoptosis, and differentiation of a variety of cell types. Patients homozygous for a null mutation of the Protein Kinase C Delta (PRKCD) gene, present clinical feature of immune dysregulation with susceptibility to Epstein-Barr virus infection. However, a minority of patients present the autoimmune lymphoproliferative syndrome (ALPS). METHODS The data were collected by direct interview and examining the patient's clinical record. Whole-exome sequencing was performed to detect the underlying genetic mutation in the patient. We also conducted electronic searches for ALPS-like reported patients in PubMed, Web of Science, and Scopus databases. RESULTS In this study, we reported a 13-year-old boy who presented with autoimmunity, lymphoproliferation, recurrent pneumonia, cardiomyopathy, and dermatological manifestations. An elevation of double-negative T cells, CD8+ T cells, serum IgG level, as well as a reduction in NK cells, was observed in the patient. A homozygous frameshift mutation (c.1293_1294insA) in exon 13 of the PRKCD gene was confirmed. The literature search showed 39 ALPS-like patients with monogenic defects which only six (15.3%) of them were due to PRKCD genes. CONCLUSION PRKCD should be considered in the context of ALPS clinical manifestations with prominent dermatological involvements.
Collapse
Affiliation(s)
- Niusha Sharifinejad
- Student Research Committee, Alborz University of Medical Sciences, Karaj, Iran.,Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Gholamreza Azizi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Nasrin Behniafard
- Department of Allergy and Clinical Immunology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Majid Zaki-Dizaji
- Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Mahnaz Jamee
- Student Research Committee, Alborz University of Medical Sciences, Karaj, Iran.,Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Reza Yazdani
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hassan Abolhassani
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Asghar Aghamohammadi
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Primary immunodeficiencies reveal the molecular requirements for effective host defense against EBV infection. Blood 2020; 135:644-655. [PMID: 31942615 DOI: 10.1182/blood.2019000928] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 10/14/2019] [Indexed: 01/22/2023] Open
Abstract
Epstein-Barr virus (EBV) is an enigma; on one hand, it infects and persists in latent form in the vast majority of the global population, causing relatively benign disease in otherwise healthy individuals. On the other hand, EBV represents the first identified oncogenic virus, capable of causing ≥7 different types of malignancies, usually in immunocompromised individuals. Furthermore, some individuals with defined inborn errors of immunity exhibit extreme susceptibility to EBV-induced disease, developing severe and often fatal infectious mononucleosis, hemophagocytic lymphohistiocytosis, lymphoproliferative disease, and/or EBV+ B-cell lymphoma. Thus, host and pathogen have coevolved to enable viral persistence and survival with minimal collateral damage to the healthy host. However, acquired or genetic disruptions to host defense that tip the balance in favor of EBV can have catastrophic effects. The study of primary immunodeficiencies has provided opportunities to define nonredundant requirements for host defense against EBV infection. This has not only revealed mechanisms underlying EBV-induced disease in these primary immunodeficiencies but also identified molecules and pathways that could be targeted to enhance the efficacy of an EBV-specific vaccine or treat severe EBV infection and pathological consequences in immunodeficient hosts.
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW Advances in genomics and animal models of human disease have enabled the discovery of mechanisms important for host immunity and self-tolerance. Here, we summarize conceptual and clinical discoveries identified from 2018 to 2019 in the field of primary immunodeficiencies and autoimmunity. RECENT FINDINGS Three new primary immunodeficiencies with autoimmunity were identified and the clinical phenotypes of NFKB1 haploinsufficiency and RASGRP1 deficiency were expanded. A diversity of novel mechanisms leading to autoimmunity associated with primary immunodeficiencies (PIDs) was reported, including pathways important for the metabolism and function of regulatory T cells and germinal B cells, the contribution of neutrophil extracellular traps to plasmacytoid dendritic cell activation and the influence of commensal bacteria on the generation of autoantibodies. With regard to therapeutic developments in the field, we highlight the use of janus kinase inhibitors for immune dysregulation associated with gain-of-function variants in STAT1 and STAT3, as well as the risks of persistent hypogammaglobulinemia associated with rituximab treatment. SUMMARY Mechanistic studies of PIDs with autoimmunity elucidate key principles governing the balance between immune surveillance and self-tolerance.
Collapse
|
25
|
Zeng T, Lv G, Chen X, Yang L, Zhou L, Dou Y, Tang X, Yang J, An Y, Zhao X. CD8 + T-cell senescence and skewed lymphocyte subsets in young Dyskeratosis Congenita patients with PARN and DKC1 mutations. J Clin Lab Anal 2020; 34:e23375. [PMID: 32452087 PMCID: PMC7521304 DOI: 10.1002/jcla.23375] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/16/2020] [Accepted: 05/01/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Dyskeratosis congenita (DC) is a syndrome resulting from defective telomere maintenance. Immunodeficiency associated with DC can cause significant morbidity and lead to premature mortality, but the immunological characteristics and molecular hallmark of DC patients, especially young patients, have not been described in detail. METHODS We summarize the clinical data of two juvenile patients with DC. Gene mutations were identified by whole-exome and direct sequencing. Swiss-PdbViewer was used to predict the pathogenicity of identified mutations. The relative telomere length was determined by QPCR, and a comprehensive analysis of lymphocyte subsets and CD57 expression was performed by flow cytometry. RESULTS Both patients showed typical features of DC without severe infection. In addition, patient 1 (P1) was diagnosed with Hoyeraal-Hreidarsson syndrome due to cerebellar hypoplasia. Gene sequencing showed P1 had a compound heterozygous mutation (c.204G > T and c.178-245del) in PARN and P2 had a novel hemizygous mutation in DKC1 (c.1051A > G). Lymphocyte subset analysis showed B and NK cytopenia, an inverted CD4:CD8 ratio, and decreased naïve CD4 and CD8 cells. A significant increase in CD21low B cells and skewed numbers of helper T cells (Th), regulatory T cells (Treg), follicular regulatory T cells (Tfr), and follicular helper T cells (Tfh) were also detected. Short telomere lengths, increased CD57 expression, and an expansion of CD8 effector memory T cells re-expressing CD45RA (TEMRA) were also found in both patients. CONCLUSION Unique immunologic abnormalities, CD8 T-cell senescence, and shortened telomere together as a hallmark occur in young DC patients before progression to severe disease.
Collapse
Affiliation(s)
- Ting Zeng
- Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing)China International Science and Technology Cooperation base of Child Development and Critical DisordersChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Child Infection and ImmunityChildren's Hospital of Chongqing Medical UniversityChongqingChina
| | - Ge Lv
- Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing)China International Science and Technology Cooperation base of Child Development and Critical DisordersChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Child Infection and ImmunityChildren's Hospital of Chongqing Medical UniversityChongqingChina
| | - Xuemei Chen
- Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing)China International Science and Technology Cooperation base of Child Development and Critical DisordersChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Child Infection and ImmunityChildren's Hospital of Chongqing Medical UniversityChongqingChina
| | - Lu Yang
- Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing)China International Science and Technology Cooperation base of Child Development and Critical DisordersChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Child Infection and ImmunityChildren's Hospital of Chongqing Medical UniversityChongqingChina
| | - Lina Zhou
- Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing)China International Science and Technology Cooperation base of Child Development and Critical DisordersChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Child Infection and ImmunityChildren's Hospital of Chongqing Medical UniversityChongqingChina
| | - Ying Dou
- Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing)China International Science and Technology Cooperation base of Child Development and Critical DisordersChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Child Infection and ImmunityChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Department of Hematology and OncologyChildren's Hospital of Chongqing Medical UniversityChongqingChina
| | - Xuemei Tang
- Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing)China International Science and Technology Cooperation base of Child Development and Critical DisordersChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Child Infection and ImmunityChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Department of Rheumatology and ImmunologyChildren's Hospital of Chongqing Medical UniversityChongqingChina
| | - Jun Yang
- Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing)China International Science and Technology Cooperation base of Child Development and Critical DisordersChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Child Infection and ImmunityChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Department of Rheumatology and ImmunologyShenzhen Children's HospitalShenzhenChina
| | - Yunfei An
- Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing)China International Science and Technology Cooperation base of Child Development and Critical DisordersChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Child Infection and ImmunityChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Department of Rheumatology and ImmunologyChildren's Hospital of Chongqing Medical UniversityChongqingChina
| | - Xiaodong Zhao
- Pediatric Research InstituteMinistry of Education Key Laboratory of Child Development and DisordersNational Clinical Research Center for Child Health and Disorders (Chongqing)China International Science and Technology Cooperation base of Child Development and Critical DisordersChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Child Infection and ImmunityChildren's Hospital of Chongqing Medical UniversityChongqingChina
- Department of Rheumatology and ImmunologyChildren's Hospital of Chongqing Medical UniversityChongqingChina
| |
Collapse
|
26
|
Khan H, Ullah H, Khattak S, Aschner M, Aguilar CN, Halimi SMA, Cauli O, Shah SMM. Therapeutic potential of alkaloids in autoimmune diseases: Promising candidates for clinical trials. Phytother Res 2020; 35:50-62. [PMID: 32667693 DOI: 10.1002/ptr.6763] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/23/2020] [Accepted: 05/16/2020] [Indexed: 02/05/2023]
Abstract
Clinical investigations have characterized numerous disorders like autoimmune diseases, affecting the population at a rate of approximately 8-10%. These disorders are characterized by T-cell and auto-antibodies responses to self-molecules by immune system reactivity. Several therapeutic options have been adopted in clinics to combat such diseases, however, most of them are recurring. Thus, the discovery of new effective agents for the treatment of autoimmune diseases is paramount. In this context, natural products might be a useful alternative to the current therapies. Plant alkaloids with their substantial therapeutic history can be particularly interesting candidates for the alleviation of autoimmune ailments. This review encompasses various alkaloids with significant effects against autoimmune diseases in preclinical trials. These results suggest further clinical assessment with respect to autoimmune illnesses. Furthermore, the application of modern technologies such as nanoformulation could be also helpful in the design of more effective therapies and thus further studies are needed to decipher their therapeutic efficacy as well as potential limitations.
Collapse
Affiliation(s)
- Haroon Khan
- Abdul Wali khan university Mardan, Abdul Wali khan university Mardan, Department of Pharmacy, Abdul Wali Khan university Mardan, Pakistan, Mardan, Pakistan, 23200, Pakistan
| | - Hammad Ullah
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Sumaira Khattak
- Abdul Wali khan university Mardan, Abdul Wali khan university Mardan, Department of Pharmacy, Abdul Wali Khan university Mardan, Pakistan, Mardan, Pakistan, 23200, Pakistan
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, New York, USA
| | - Cristobal N Aguilar
- School of Chemistry, Universidad Autónoma de Coahuila Saltillo, Saltillo, Mexico
| | - Syed M A Halimi
- Department of Pharmacy, University of Peshawar, Peshawar, Pakistan
| | - Omar Cauli
- Department of Nursing, University of Valencia, Valencia, Spain
| | - Syed M M Shah
- Department of Pharmacy, University of Swabi Pakistan, Swabi, Pakistan
| |
Collapse
|
27
|
Sharma T, Cotney J, Singh V, Sanjay A, Reichenberger EJ, Ueki Y, Maye P. Investigating global gene expression changes in a murine model of cherubism. Bone 2020; 135:115315. [PMID: 32165349 PMCID: PMC7305689 DOI: 10.1016/j.bone.2020.115315] [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: 12/26/2019] [Revised: 02/25/2020] [Accepted: 03/08/2020] [Indexed: 11/22/2022]
Abstract
Cherubism is a rare genetic disorder caused primarily by mutations in SH3BP2 resulting in excessive bone resorption and fibrous tissue overgrowth in the lower portions of the face. Bone marrow derived cell cultures derived from a murine model of cherubism display poor osteogenesis and spontaneous osteoclast formation. To develop a deeper understanding for the potential underlying mechanisms contributing to these phenotypes in mice, we compared global gene expression changes in hematopoietic and mesenchymal cell populations between cherubism and wild type mice. In the hematopoietic population, not surprisingly, upregulated genes were significantly enriched for functions related to osteoclastogenesis. However, these upregulated genes were also significantly enriched for functions associated with inflammation including arachidonic acid/prostaglandin signaling, regulators of coagulation and autoinflammation, extracellular matrix remodeling, and chemokine expression. In the mesenchymal population, we observed down regulation of osteoblast and adventitial reticular cell marker genes. Regulators of BMP and Wnt pathway associated genes showed numerous changes in gene expression, likely implicating the down regulation of BMP signaling and possibly the activation of certain Wnt pathways. Analyses of the cherubism derived mesenchymal population also revealed interesting changes in gene expression related to inflammation including the expression of distinct granzymes, chemokines, and sulfotransferases. These studies reveal complex changes in gene expression elicited from a cherubic mutation in Sh3bp2 that are informative to the mechanisms responding to inflammatory stimuli and repressing osteogenesis. The outcomes of this work are likely to have relevance not only to cherubism, but other inflammatory conditions impacting the skeleton.
Collapse
Affiliation(s)
- Tulika Sharma
- Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health, United States of America
| | - Justin Cotney
- Department of Genetics and Genome Sciences, University of Connecticut Health, United States of America
| | - Vijender Singh
- Computational Biology Core, Institute for Systems Genomics, University of Connecticut, United States of America
| | - Archana Sanjay
- Department of Orthopedic Surgery, University of Connecticut Health, United States of America
| | - Ernst J Reichenberger
- Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health, United States of America
| | - Yasuyoshi Ueki
- Department of Biomedical Sciences and Comprehensive Care, Indiana University, United States of America
| | - Peter Maye
- Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health, United States of America.
| |
Collapse
|
28
|
Tangye SG. Genetic susceptibility to EBV infection: insights from inborn errors of immunity. Hum Genet 2020; 139:885-901. [PMID: 32152698 DOI: 10.1007/s00439-020-02145-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 02/27/2020] [Indexed: 02/07/2023]
Abstract
Epstein-Barr virus (EBV) is a ubiquitous human pathogen, infecting > 90% of the adult population. In the vast majority of healthy individuals, infection with EBV runs a relatively benign course. However, EBV is by no means a benign pathogen. Indeed, apart from being associated with at least seven different types of malignancies, EBV infection can cause severe and often fatal diseases-hemophagocytic lymphohistiocytosis, lymphoproliferative disease, B-cell lymphoma-in rare individuals with specific monogenic inborn errors of immunity. The discovery and detailed investigation of inborn errors of immunity characterized by heightened susceptibility to, or increased frequency of, EBV-induced disease have elegantly revealed cell types and signaling pathways that play critical and non-redundant roles in host-defense against EBV. These analyses have revealed not only mechanisms underlying EBV-induced disease in rare genetic conditions, but also identified molecules and pathways that could be targeted to treat severe EBV infection and pathological consequences in immunodeficient hosts, or even potentially enhance the efficacy of an EBV-specific vaccine.
Collapse
Affiliation(s)
- Stuart G Tangye
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2010, Australia. .,St. Vincent's Clinical School, University of NSW Sydney, Darlinghurst, NSW, 2010, Australia. .,Clincial Immunogenomics Research Consortium Australasia (CIRCA), Darlinghurst, NSW, Australia.
| |
Collapse
|
29
|
Latour S, Fischer A. Signaling pathways involved in the T-cell-mediated immunity against Epstein-Barr virus: Lessons from genetic diseases. Immunol Rev 2020; 291:174-189. [PMID: 31402499 DOI: 10.1111/imr.12791] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/05/2019] [Accepted: 06/10/2019] [Indexed: 12/13/2022]
Abstract
Primary immunodeficiencies (PIDs) provide researchers with unique models to understand in vivo immune responses in general and immunity to infections in particular. In humans, impaired immune control of Epstein-Barr virus (EBV) infection is associated with the occurrence of several different immunopathologic conditions; these include non-malignant and malignant B-cell lymphoproliferative disorders, hemophagocytic lymphohistiocytosis (HLH), a severe inflammatory condition, and a chronic acute EBV infection of T cells. Studies of PIDs associated with a predisposition to develop severe, chronic EBV infections have led to the identification of key components of immunity to EBV - notably the central role of T-cell expansion and its regulation in the pathophysiology of EBV-associated diseases. On one hand, the defective expansion of EBV-specific CD8 T cells results from mutations in genes involved in T-cell activation (such as RASGRP1, MAGT1, and ITK), DNA metabolism (CTPS1) or co-stimulatory pathways (CD70, CD27, and TNFSFR9 (also known as CD137/4-1BB)) leads to impaired elimination of proliferating EBV-infected B cells and the occurrence of lymphoma. On the other hand, protracted T-cell expansion and activation after the defective killing of EBV-infected B cells is caused by genetic defects in the components of the lytic granule exocytosis pathway or in the small adapter protein SH2D1A (also known as SAP), a key activator of T- and NK cell-cytotoxicity. In this setting, the persistence of EBV-infected cells results in HLH, a condition characterized by unleashed T-cell and macrophage activation. Moreover, genetic defects causing selective vulnerability to EBV infection have highlighted the role of co-receptor molecules (CD27, CD137, and SLAM-R) selectively involved in immune responses against infected B cells via specific T-B cell interactions.
Collapse
Affiliation(s)
- Sylvain Latour
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, Inserm UMR 1163, Paris, France.,University Paris Descartes Sorbonne Paris Cité, Imagine Institut, Paris, France
| | - Alain Fischer
- University Paris Descartes Sorbonne Paris Cité, Imagine Institut, Paris, France.,Department of Pediatric Immunology, Hematology and Rheumatology, Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Paris, France.,Collège de France, Paris, France.,Inserm UMR 1163, Paris, France
| |
Collapse
|
30
|
Wasik U, Kempinska-Podhorodecka A, Bogdanos DP, Milkiewicz P, Milkiewicz M. Enhanced expression of miR-21 and miR-150 is a feature of anti-mitochondrial antibody-negative primary biliary cholangitis. Mol Med 2020; 26:8. [PMID: 31948396 PMCID: PMC6966805 DOI: 10.1186/s10020-019-0130-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 12/17/2019] [Indexed: 02/02/2023] Open
Abstract
Background & Aims Anti-mitochondrial-autoantibodies (AMA) remain a hallmark of Primary Biliary Cholangitis (PBC) however approximately 10% of patients test negative for these antibodies. They do not differ in terms of biochemistry or clinical presentation from AMA positive ones. Epigenetics play a key role in immune signalling. Two microRNAs (miRs), namely, miR-21 and miR-150 are known to be involved in liver inflammation and fibrosis. The expression of those two microRNAs and their downstream targets were analyze in the context of AMA-status and the stage of liver fibrosis. Methods The relative levels of miR-21 and miR-150 and their target genes: cMyb, RAS-guanyl-releasing protein-1(RASGRP1), and DNA-methyltransferase-1(DNMT1) were determined by Real-Time PCR in serum, liver tissue and peripheral blood mononuclear cells (PBMCs) of patients with PBC. Results Serum expressions of miR-21 and miR-150 were significantly enhanced in AMA-negative patients, and they inversely correlated with disease-specific AMA titers in PBS patients. In PBMCs, an increased expression of miR-21 correlated with decreased levels of RASGRP1 and DNMT1 mRNAs whereas, the level of miR-150 remained comparable to controls; and cMyb mRNA was downregulated. In cirrhotic livers, the level of miR-21 was unchanged while miR-150 expression was increased. Conclusion This study convincingly report, that AMA-negative PBC is characterized by notable alternations of miR-21 and miR-150 and their downstream targets compared to AMA-positive patients underlining their possible importance in the induction of the disease and its progression to fibrosis.
Collapse
Affiliation(s)
- Urszula Wasik
- Department of Medical Biology, Pomeranian Medical University, Szczecin, Poland
| | | | - Dimitrios P Bogdanos
- Department of Rheumatology and Clinical Immunology, University of Thessaly, Larissa, Greece
| | - Piotr Milkiewicz
- Translational Medicine Group, Pomeranian Medical University, Szczecin, Poland.,Liver and Internal Medicine Unit, Medical University of Warsaw, Warsaw, Poland
| | | |
Collapse
|
31
|
Cabral-Marques O, Schimke LF, de Oliveira EB, El Khawanky N, Ramos RN, Al-Ramadi BK, Segundo GRS, Ochs HD, Condino-Neto A. Flow Cytometry Contributions for the Diagnosis and Immunopathological Characterization of Primary Immunodeficiency Diseases With Immune Dysregulation. Front Immunol 2019; 10:2742. [PMID: 31849949 PMCID: PMC6889851 DOI: 10.3389/fimmu.2019.02742] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 11/08/2019] [Indexed: 12/24/2022] Open
Abstract
Almost 70 years after establishing the concept of primary immunodeficiency disorders (PIDs), more than 320 monogenic inborn errors of immunity have been identified thanks to the remarkable contribution of high-throughput genetic screening in the last decade. Approximately 40 of these PIDs present with autoimmune or auto-inflammatory symptoms as the primary clinical manifestation instead of infections. These PIDs are now recognized as diseases of immune dysregulation. Loss-of function mutations in genes such as FOXP3, CD25, LRBA, IL-10, IL10RA, and IL10RB, as well as heterozygous gain-of-function mutations in JAK1 and STAT3 have been reported as causative of these disorders. Identifying these syndromes has considerably contributed to expanding our knowledge on the mechanisms of immune regulation and tolerance. Although whole exome and whole genome sequencing have been extremely useful in identifying novel causative genes underlying new phenotypes, these approaches are time-consuming and expensive. Patients with monogenic syndromes associated with autoimmunity require faster diagnostic tools to delineate therapeutic strategies and avoid organ damage. Since these PIDs present with severe life-threatening phenotypes, the need for a precise diagnosis in order to initiate appropriate patient management is necessary. More traditional approaches such as flow cytometry are therefore a valid option. Here, we review the application of flow cytometry and discuss the relevance of this powerful technique in diagnosing patients with PIDs presenting with immune dysregulation. In addition, flow cytometry represents a fast, robust, and sensitive approach that efficiently uncovers new immunopathological mechanisms underlying monogenic PIDs.
Collapse
Affiliation(s)
- Otavio Cabral-Marques
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Lena F Schimke
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, University of Freiburg, Freiburg im Breisgau, Germany
| | | | - Nadia El Khawanky
- Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg University Medical Center, Freiburg im Breisgau, Germany.,Precision Medicine Theme, The South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Rodrigo Nalio Ramos
- INSERM U932, SiRIC Translational Immunotherapy Team, Institut Curie, Paris Sciences et Lettres Research University, Paris, France
| | - Basel K Al-Ramadi
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | | | - Hans D Ochs
- Department of Pediatrics, University of Washington School of Medicine, and Seattle Children's Research Institute, Seattle, WA, United States
| | - Antonio Condino-Neto
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
32
|
Lazy Leukocyte Syndrome-an Enigma Finally Solved? J Clin Immunol 2019; 40:9-12. [PMID: 31768891 DOI: 10.1007/s10875-019-00718-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 11/04/2019] [Indexed: 01/09/2023]
|
33
|
Molineros JE, Singh B, Terao C, Okada Y, Kaplan J, McDaniel B, Akizuki S, Sun C, Webb CF, Looger LL, Nath SK. Mechanistic Characterization of RASGRP1 Variants Identifies an hnRNP-K-Regulated Transcriptional Enhancer Contributing to SLE Susceptibility. Front Immunol 2019; 10:1066. [PMID: 31164884 PMCID: PMC6536009 DOI: 10.3389/fimmu.2019.01066] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 04/25/2019] [Indexed: 11/21/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease with a strong genetic component. We recently identified a novel SLE susceptibility locus near RASGRP1, which governs the ERK/MAPK kinase cascade and B-/T-cell differentiation and development. However, precise causal RASGRP1 functional variant(s) and their mechanisms of action in SLE pathogenesis remain undefined. Our goal was to fine-map this locus, prioritize genetic variants likely to be functional, experimentally validate their biochemical mechanisms, and determine the contribution of these SNPs to SLE risk. We performed a meta-analysis across six Asian and European cohorts (9,529 cases; 22,462 controls), followed by in silico bioinformatic and epigenetic analyses to prioritize potentially functional SNPs. We experimentally validated the functional significance and mechanism of action of three SNPs in cultured T-cells. Meta-analysis identified 18 genome-wide significant (p < 5 × 10−8) SNPs, mostly concentrated in two haplotype blocks, one intronic and the other intergenic. Epigenetic fine-mapping, allelic, eQTL, and imbalance analyses predicted three transcriptional regulatory regions with four SNPs (rs7170151, rs11631591-rs7173565, and rs9920715) prioritized for functional validation. Luciferase reporter assays indicated significant allele-specific enhancer activity for intronic rs7170151 and rs11631591-rs7173565 in T-lymphoid (Jurkat) cells, but not in HEK293 cells. Following up with EMSA, mass spectrometry, and ChIP-qPCR, we detected allele-dependent interactions between heterogeneous nuclear ribonucleoprotein K (hnRNP-K) and rs11631591. Furthermore, inhibition of hnRNP-K in Jurkat and primary T-cells downregulated RASGRP1 and ERK/MAPK signaling. Comprehensive association, bioinformatics, and epigenetic analyses yielded putative functional variants of RASGRP1, which were experimentally validated. Notably, intronic variant (rs11631591) is located in a cell type-specific enhancer sequence, where its risk allele binds to the hnRNP-K protein and modulates RASGRP1 expression in Jurkat and primary T-cells. As risk allele dosage of rs11631591 correlates with increased RASGRP1 expression and ERK activity, we suggest that this SNP may underlie SLE risk at this locus.
Collapse
Affiliation(s)
- Julio E Molineros
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Bhupinder Singh
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Chikashi Terao
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Jakub Kaplan
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Barbara McDaniel
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Shuji Akizuki
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Celi Sun
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Carol F Webb
- Departments of Medicine, Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Loren L Looger
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA, United States
| | - Swapan K Nath
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| |
Collapse
|
34
|
Kono M, Kurita T, Yasuda S, Kono M, Fujieda Y, Bohgaki T, Katsuyama T, Tsokos GC, Moulton VR, Atsumi T. Decreased Expression of Serine/Arginine-Rich Splicing Factor 1 in T Cells From Patients With Active Systemic Lupus Erythematosus Accounts for Reduced Expression of RasGRP1 and DNA Methyltransferase 1. Arthritis Rheumatol 2018; 70:2046-2056. [PMID: 29905030 DOI: 10.1002/art.40585] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 06/07/2018] [Indexed: 02/03/2023]
Abstract
OBJECTIVE T cells from systemic lupus erythematosus (SLE) patients have reduced protein levels of RasGRP1, a guanine nucleotide exchange factor for Ras, and increased transcript of alternatively spliced (AS) forms lacking exon 11. Serine/arginine-rich splicing factor 1 (SRSF1) binds pre-messenger RNA (pre-mRNA) to regulate AS forms of several genes, including CD3ζ in SLE T cells. This study was undertaken to assess whether SRSF1 controls the expression of RasGRP1 in T cells from patients with SLE. METHODS We studied T cells from 45 SLE patients and 18 healthy subjects. Expression levels of SRSF1, wild-type (WT) RasGRP1, and DNA methyltransferase 1 (DNMT1) were assessed by quantitative polymerase chain reaction. Direct binding of SRSF1 to exon 11 of RasGRP1 mRNA was evaluated with an oligonucleotide-protein pulldown assay. Healthy T cells and SLE T cells were treated with SRSF1-specific small interfering RNA or SRSF1 expression vector, respectively, and then evaluated for mRNA/protein expression. RESULTS SRSF1 expression levels were significantly lower in T cells from SLE patients compared to those from healthy subjects, and correlated inversely with disease activity and positively with levels of RasGRP1-WT and DNMT1. SRSF1 bound directly to exon 11 of RasGRP1 mRNA. Silencing of SRSF1 in human T cells led to increased ratios of RasGRP1-AS to RasGRP1-WT and decreased levels of RasGRP1 protein, whereas overexpression of SRSF1 in SLE T cells caused recovery of RasGRP1, which in turn induced DNMT1/interleukin-2 expression. CONCLUSION SRSF1 controls the alternative splicing of RasGRP1 and subsequent protein expression. Our findings extend evidence that alternative splicing plays a central role in the aberrant T cell function in patients with SLE by controlling the expression of multiple genes.
Collapse
Affiliation(s)
| | | | | | - Michihito Kono
- Hokkaido University, Sapporo, Japan, and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | | | | | - Takayuki Katsuyama
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - George C Tsokos
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Vaishali R Moulton
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | | |
Collapse
|
35
|
Somekh I, Marquardt B, Liu Y, Rohlfs M, Hollizeck S, Karakukcu M, Unal E, Yilmaz E, Patiroglu T, Cansever M, Frizinsky S, Vishnvenska-Dai V, Rechavi E, Stauber T, Simon AJ, Lev A, Klein C, Kotlarz D, Somech R. Novel Mutations in RASGRP1 are Associated with Immunodeficiency, Immune Dysregulation, and EBV-Induced Lymphoma. J Clin Immunol 2018; 38:699-710. [DOI: 10.1007/s10875-018-0533-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 07/09/2018] [Indexed: 12/25/2022]
|
36
|
Latour S, Winter S. Inherited Immunodeficiencies With High Predisposition to Epstein-Barr Virus-Driven Lymphoproliferative Diseases. Front Immunol 2018; 9:1103. [PMID: 29942301 PMCID: PMC6004768 DOI: 10.3389/fimmu.2018.01103] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 05/02/2018] [Indexed: 01/16/2023] Open
Abstract
Epstein–Barr Virus (EBV) is a gamma-herpes virus that infects 90% of humans without any symptoms in most cases, but has an oncogenic potential, especially in immunocompromised individuals. In the past 30 years, several primary immunodeficiencies (PIDs) associated with a high risk to develop EBV-associated lymphoproliferative disorders (LPDs), essentially consisting of virus-associated hemophagocytic syndrome, non-malignant and malignant B-cell LPDs including non-Hodgkin and Hodgkin’s types of B lymphomas have been characterized. Among them are SH2D1A (SAP), XIAP, ITK, MAGT1, CD27, CD70, CTPS1, RASGRP1, and CORO1A deficiencies. Penetrance of EBV infection ranges from 50 to 100% in those PIDs. Description of large cohorts and case reports has refined the specific phenotypes associated with these PIDs helping to the diagnosis. Specific pathways required for protective immunity to EBV have emerged from studies of these PIDs. SLAM-associated protein-dependent SLAM receptors and MAGT1-dependent NKG2D pathways are important for T and NK-cell cytotoxicity toward EBV-infected B-cells, while CD27–CD70 interactions are critical to drive the expansion of EBV-specific T-cells. CTPS1 and RASGRP1 deficiencies further strengthen that T-lymphocyte expansion is a key step in the immune response to EBV. These pathways appear to be also important for the anti-tumoral immune surveillance of abnormal B cells. Monogenic PIDs should be thus considered in case of any EBV-associated LPDs.
Collapse
Affiliation(s)
- Sylvain Latour
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Paris, France.,Imagine Institute, Paris Descartes University, Sorbonne Paris Cité, Paris, France.,Equipe de Recherche Labéllisée, Ligue National contre le Cancer, Paris, France
| | - Sarah Winter
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Paris, France.,Imagine Institute, Paris Descartes University, Sorbonne Paris Cité, Paris, France.,Equipe de Recherche Labéllisée, Ligue National contre le Cancer, Paris, France
| |
Collapse
|
37
|
Abstract
Proper regulation of the immune system is required for protection against pathogens and preventing autoimmune disorders. Inborn errors of the immune system due to inherited or de novo germline mutations can lead to the loss of protective immunity, aberrant immune homeostasis, and the development of autoimmune disease, or combinations of these. Forward genetic screens involving clinical material from patients with primary immunodeficiencies (PIDs) can vary in severity from life-threatening disease affecting multiple cell types and organs to relatively mild disease with susceptibility to a limited range of pathogens or mild autoimmune conditions. As central mediators of innate and adaptive immune responses, T cells are critical orchestrators and effectors of the immune response. As such, several PIDs result from loss of or altered T cell function. PID-associated functional defects range from complete absence of T cell development to uncontrolled effector cell activation. Furthermore, the gene products of known PID causal genes are involved in diverse molecular pathways ranging from T cell receptor signaling to regulators of protein glycosylation. Identification of the molecular and biochemical cause of PIDs can not only guide the course of treatment for patients, but also inform our understanding of the basic biology behind T cell function. In this chapter, we review PIDs with known genetic causes that intrinsically affect T cell function with particular focus on perturbations of biochemical pathways.
Collapse
Affiliation(s)
- William A Comrie
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States; Clinical Genomics Program, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, United States
| | - Michael J Lenardo
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States; Clinical Genomics Program, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, United States.
| |
Collapse
|