1
|
Consonni F, Moreno S, Vinuales Colell B, Stolzenberg MC, Fernandes A, Parisot M, Masson C, Neveux N, Rosain J, Bamberger S, Vigue MG, Malphettes M, Quartier P, Picard C, Rieux-Laucat F, Magerus A. Study of the potential role of CASPASE-10 mutations in the development of autoimmune lymphoproliferative syndrome. Cell Death Dis 2024; 15:315. [PMID: 38704374 PMCID: PMC11069523 DOI: 10.1038/s41419-024-06679-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 04/09/2024] [Accepted: 04/12/2024] [Indexed: 05/06/2024]
Abstract
Autoimmune lymphoproliferative syndrome (ALPS) is a primary disorder of lymphocyte homeostasis, leading to chronic lymphoproliferation, autoimmune cytopenia, and increased risk of lymphoma. The genetic landscape of ALPS includes mutations in FAS, FASLG, and FADD, all associated with apoptosis deficiency, while the role of CASP10 defect in the disease remains debated. In this study, we aimed to assess the impact of CASP10 variants on ALPS pathogenesis. We benefit from thousands of genetic analysis datasets performed in our Institute's genetic platform to identify individuals carrying CASP10 variants previously suspected to be involved in ALPS outcome: p.C401LfsX15, p.V410I and p.Y446C, both at heterozygous and homozygous state. Clinical and laboratory features of the six included subjects were variable but not consistent with ALPS. Two individuals were healthy. Comprehensive analyses of CASP10 protein expression and FAS-mediated apoptosis were conducted and compared to healthy controls and ALPS patients with FAS mutations. Missense CASP10 variants (p.V410I and p.Y446C), which are common in the general population, did not disrupt CASP10 expression, nor FAS-mediated apoptosis. In contrast, homozygous p.C401LfsX15 CASP10 variant lead to a complete abolished CASP10 expression but had no impact on FAS-mediated apoptosis function. At heterozygous state, this p.C401LfsX15 variant lead to a reduced CASP10 protein levels but remained associated with a normal FAS-mediated apoptosis function. These findings demonstrate that CASPASE 10 is dispensable for FAS-mediated apoptosis. In consequences, CASP10 defect unlikely contribute to ALPS pathogenesis, since they did not result in an impairment of FAS-mediated apoptosis nor in clinical features of ALPS in human. Moreover, the absence of FAS expression up-regulation in subjects with CASP10 variants rule out any compensatory mechanisms possibly involved in the normal apoptosis function observed. In conclusion, this study challenges the notion that CASP10 variants contribute to the development of ALPS.
Collapse
Affiliation(s)
- Filippo Consonni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
- Centre of Excellence, Division of Paediatric Oncology/Haematology, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Solange Moreno
- University of Paris Cité, Paris, France
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Blanca Vinuales Colell
- University of Paris Cité, Paris, France
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Marie-Claude Stolzenberg
- University of Paris Cité, Paris, France
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Alicia Fernandes
- Plateforme Vecteurs Viraux et Transfert de Gènes, SFR Necker, INSERM US 24/CNRS UAR 3633, Faculté de santé Necker, Paris, France
| | - Mélanie Parisot
- University of Paris Cité, Paris, France
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163 et INSERM US24/CNRS UAR3633, Paris, France
| | - Cécile Masson
- University of Paris Cité, Paris, France
- Bioinformatics Core Facility, Paris-Cité University-Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Paris, France
| | - Nathalie Neveux
- Laboratory of Biological Nutrition, EA 4466, Faculty of Pharmacy, Paris University, Paris, France
- Clinical Chemistry Department, Hôpital Cochin, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Jérémie Rosain
- University of Paris Cité, Paris, France
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Center for the Study of Primary Immunodeficiencies, Necker Hospital for Sick Children, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Sarah Bamberger
- Pediatrics Gastroenterology and Nutrition, Robert-Debré Hospital, Paris, France
| | - Marie-Gabrielle Vigue
- Pediatrics, Infectiology, Rhumatology, Hôpital Arnaud-de-Villeneuve, CHRU de Montpellier, Montpellier, France
| | - Marion Malphettes
- University of Paris Cité, Paris, France
- Department of Clinical Immunology, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Pierre Quartier
- University of Paris Cité, Paris, France
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, INSERM UMR 1163, Paris, France
- Pediatric immuno-hematology and rheumatology department, Necker-Enfants Malades Hospital, Assistance publique - Hôpitaux de Paris, Paris, France
| | - Capucine Picard
- University of Paris Cité, Paris, France
- Center for the Study of Primary Immunodeficiencies, Necker Hospital for Sick Children, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- Pediatric immuno-hematology and rheumatology department, Necker-Enfants Malades Hospital, Assistance publique - Hôpitaux de Paris, Paris, France
| | - Frédéric Rieux-Laucat
- University of Paris Cité, Paris, France
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Aude Magerus
- University of Paris Cité, Paris, France.
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, INSERM UMR 1163, Paris, France.
| |
Collapse
|
2
|
Fernandez I, Touzot F. Looking for ALPS: The value of a combined assessment of biochemical markers. Pediatr Allergy Immunol 2024; 35:e14135. [PMID: 38700373 DOI: 10.1111/pai.14135] [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: 03/01/2024] [Revised: 04/12/2024] [Accepted: 04/16/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND Autoimmune lymphoproliferative syndrome (ALPS) is a rare primary immune disorder caused by defect of the extrinsic apoptotic pathway. The current diagnostic criteria combine clinical features and typical biomarkers but have not been the object of clear international consensus. METHODS We conducted a retrospective study on pediatric patients who were investigated for autoimmune cytopenia and/or lymphoproliferation at the CHU Sainte-Justine Hospital over 10 years. Patients were screened using the combination of TCRαβ+ CD4- CD8- "double negative" (DN) T cells and soluble plasmatic FAS ligand (sFASL). RESULTS Among the 398 tested patients, the median sFASL and DN T cells were 200 ng/mL and 1.8% of TCRαβ+ T cells, respectively. sFASL was highly correlated with vitamin B12 levels. We identified five patients diagnosed with ALPS for whose sFASL and vitamin B12 levels were the more discriminating biomarkers. While ALPS diagnostic criteria had high sensibility, their predictive value remained low. CONCLUSION sFASL level can efficiently discriminate patients with ALPS when using the appropriate thresholds. Our study highlights the need for an international consensus to redefine the place and threshold of biological biomarkers for ALPS diagnosis.
Collapse
Affiliation(s)
- Isabel Fernandez
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montréal, Québec, Canada
| | - Fabien Touzot
- Department of Pediatrics, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
3
|
Magerus A, Rensing-Ehl A, Rao VK, Teachey DT, Rieux-Laucat F, Ehl S. Autoimmune lymphoproliferative immunodeficiencies (ALPIDs): A proposed approach to redefining ALPS and other lymphoproliferative immune disorders. J Allergy Clin Immunol 2024; 153:67-76. [PMID: 37977527 PMCID: PMC10841637 DOI: 10.1016/j.jaci.2023.11.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/10/2023] [Accepted: 11/14/2023] [Indexed: 11/19/2023]
Abstract
Chronic nonmalignant lymphoproliferation and autoimmune cytopenia are relevant manifestations of immunohematologic diseases of childhood. Their diagnostic classification is challenging but important for therapy. Autoimmune lymphoproliferative syndrome (ALPS) is a genetically defined inborn error of immunity combining these manifestations, but it can explain only a small proportion of cases. Diagnostic categories such as ALPS-like disease, common variable immunodeficiency, or Evans syndrome have therefore been used. Advances in genetics and increasing availablity of targeted therapies call for more therapy-oriented disease classification. Moreover, recent discoveries in the (re)analysis of genetic conditions affecting FAS signaling ask for a more precise definition of ALPS. In this review, we propose the term autoimmune lymphoproliferative immunodeficiencies for a disease phenotype that is enriched for patients with genetic diseases for which targeted therapies are available. For patients without a current molecular diagnosis, this term defines a subgroup of immune dysregulatory disorders for further studies. Within the concept of autoimmune lymphoproliferative immunodeficiencies, we propose a revision of the ALPS classification, restricting use of this term to conditions with clear evidence of perturbation of FAS signaling and resulting specific biologic and clinical consequences. This proposed approach to redefining ALPS and other lymphoproliferative conditions provides a framework for disease classification and diagnosis that is relevant for the many specialists confronted with these diseases.
Collapse
Affiliation(s)
- Aude Magerus
- University of Paris Cité, Paris, France; Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Anne Rensing-Ehl
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - V Koneti Rao
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, Md
| | - David T Teachey
- Division of Hematology, The Children's Hospital of Philadelphia, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pa; Division of Oncology, The Children's Hospital of Philadelphia, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pa
| | - Frederic Rieux-Laucat
- University of Paris Cité, Paris, France; Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Stephan Ehl
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
4
|
Paskiewicz A, Niu J, Chang C. Autoimmune lymphoproliferative syndrome: A disorder of immune dysregulation. Autoimmun Rev 2023; 22:103442. [PMID: 37683818 DOI: 10.1016/j.autrev.2023.103442] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 09/03/2023] [Indexed: 09/10/2023]
Abstract
Autoimmune Lymphoproliferative Syndrome (ALPS) is an autoimmune disease that has been reported in over 2200 patients. It is a rare, genetic disease where pathogenic variants occur in the extrinsic pathway of apoptosis. Various mutations in different genes, such as FAS, FASL, and CASP10, can result in ALPS. Most commonly, pathogenic variants occur in the FAS receptor. This malfunctioning pathway allows for the abnormal accumulation of lymphocytes, namely CD3 + TCRαβ+CD4 - CD8- (double negative (DN) T) cells, which are a hallmark of the disease. This disease usually presents in childhood with lymphadenopathy and splenomegaly as a result of lymphoproliferation. Over time, these patients may develop cytopenias or lymphomas because of irregularities in the immune system. Current treatments include glucocorticoids, mycophenolate mofetil, sirolimus, immunoglobulin G, and rituximab. These medications serve to manage the symptoms and there are no standardized recommendations for the management of ALPS. The only curative therapy is a bone marrow transplant, but this is rarely done because of the complications. This review serves to broaden the understanding of ALPS by discussing the mechanism of immune dysregulation, how the symptoms manifest, and the mechanisms of treatment. Additionally, we discuss the epidemiology, comorbidities, and medications relating to ALPS patients across the United States using data from Cosmos.
Collapse
Affiliation(s)
- Amy Paskiewicz
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA.
| | - Jianli Niu
- Office of Human Research, Memorial Healthcare System, Hollywood, FL 33021, USA.
| | - Christopher Chang
- Division of Immunology, Allergy and Pediatric Rheumatology, Joe DiMaggio Children's Hospital, Memorial Healthcare System, Hollywood, FL 33021, USA.
| |
Collapse
|
5
|
Toskov V, Ehl S. Autoimmune lymphoproliferative immunodeficiencies (ALPID) in childhood: breakdown of immune homeostasis and immune dysregulation. Mol Cell Pediatr 2023; 10:11. [PMID: 37702894 PMCID: PMC10499775 DOI: 10.1186/s40348-023-00167-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 09/05/2023] [Indexed: 09/14/2023] Open
Abstract
Many inborn errors of immunity (IEI) manifest with hallmarks of both immunodeficiency and immune dysregulation due to uncontrolled immune responses and impaired immune homeostasis. A subgroup of these disorders frequently presents with autoimmunity and lymphoproliferation (ALPID phenotype). After the initial description of the genetic basis of autoimmune lymphoproliferative syndrome (ALPS) more than 20 years ago, progress in genetics has helped to identify many more genetic conditions underlying this ALPID phenotype. Among these, the majority is caused by a group of autosomal-dominant conditions including CTLA-4 haploinsufficiency, STAT3 gain-of-function disease, activated PI3 kinase syndrome, and NF-κB1 haploinsufficiency. Even within a defined genetic condition, ALPID patients may present with staggering clinical heterogeneity, which makes diagnosis and management a challenge. In this review, we discuss the pathophysiology, clinical presentation, approaches to diagnosis, and conventional as well as targeted therapy of the most common ALPID conditions.
Collapse
Affiliation(s)
- Vasil Toskov
- Centre for Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stephan Ehl
- Centre for Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
6
|
Mukvich OM, Telegeev GD, Matskevych AM, Gilfanova AM. Polymorphisms of Genes Associated with Intracellular Signaling Pathways in Juvenile Idiopathic Arthritis. CYTOL GENET+ 2022. [DOI: 10.3103/s0095452722030070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
7
|
Delgadillo DM, Céspedes-Cruz AI, Ríos-Castro E, Rodríguez Maldonado MG, López-Nogueda M, Márquez-Gutiérrez M, Villalobos-Manzo R, Ramírez-Reyes L, Domínguez-Fuentes M, Tapia-Ramírez J. Differential Expression of Proteins in an Atypical Presentation of Autoimmune Lymphoproliferative Syndrome. Int J Mol Sci 2022; 23:5366. [PMID: 35628184 PMCID: PMC9140392 DOI: 10.3390/ijms23105366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/02/2022] [Accepted: 05/03/2022] [Indexed: 02/04/2023] Open
Abstract
Autoimmune lymphoproliferative syndrome (ALPS) is a rare disease defined as a defect in the lymphocyte apoptotic pathway. Currently, the diagnosis of ALPS is based on clinical aspects, defective lymphocyte apoptosis and mutations in Fas, FasL and Casp 10 genes. Despite this, ALPS has been misdiagnosed. The aim of this work was to go one step further in the knowledge of the disease, through a molecular and proteomic analysis of peripheral blood mononuclear cells (PBMCs) from two children, a 13-year-old girl and a 6-year-old boy, called patient 1 and patient 2, respectively, with clinical data supporting the diagnosis of ALPS. Fas, FasL and Casp10 genes from both patients were sequenced, and a sample of the total proteins from patient 1 was analyzed by label-free proteomics. Pathway analysis of deregulated proteins from PBMCs was performed on the STRING and PANTHER bioinformatics databases. A mutation resulting in an in-frame premature stop codon and protein truncation was detected in the Fas gene from patient 2. From patient 1, the proteomic analysis showed differences in the level of expression of proteins involved in, among other processes, cell cycle, regulation of cell cycle arrest and immune response. Noticeably, the most down-regulated protein is an important regulator of the cell cycle process. This could be an explanation of the disease in patient 1.
Collapse
Affiliation(s)
- Dulce María Delgadillo
- Unidad de Genómica, Proteómica y Metabolómica, Laboratorio Nacional de Servicios Experimentales (LaNSE), Centro de Investigación y de Estudios Avanzados, Mexico City 07360, CP, Mexico
| | - Adriana Ivonne Céspedes-Cruz
- Unidad Médica de Alta Especialidad (UMAE), Centro Médico Nacional La Raza Hospital General, Mexico City 02990, CP, Mexico
| | - Emmanuel Ríos-Castro
- Unidad de Genómica, Proteómica y Metabolómica, Laboratorio Nacional de Servicios Experimentales (LaNSE), Centro de Investigación y de Estudios Avanzados, Mexico City 07360, CP, Mexico
| | | | - Mariel López-Nogueda
- Unidad Médica de Alta Especialidad (UMAE), Centro Médico Nacional La Raza Hospital General, Mexico City 02990, CP, Mexico
| | - Miguel Márquez-Gutiérrez
- Unidad Médica de Alta Especialidad (UMAE), Centro Médico Nacional La Raza Hospital General, Mexico City 02990, CP, Mexico
| | - Rocío Villalobos-Manzo
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados, Mexico City 07360, CP, Mexico
| | - Lorena Ramírez-Reyes
- Unidad de Genómica, Proteómica y Metabolómica, Laboratorio Nacional de Servicios Experimentales (LaNSE), Centro de Investigación y de Estudios Avanzados, Mexico City 07360, CP, Mexico
| | - Misael Domínguez-Fuentes
- Unidad de Genómica, Proteómica y Metabolómica, Laboratorio Nacional de Servicios Experimentales (LaNSE), Centro de Investigación y de Estudios Avanzados, Mexico City 07360, CP, Mexico
| | - José Tapia-Ramírez
- Unidad de Genómica, Proteómica y Metabolómica, Laboratorio Nacional de Servicios Experimentales (LaNSE), Centro de Investigación y de Estudios Avanzados, Mexico City 07360, CP, Mexico
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados, Mexico City 07360, CP, Mexico
| |
Collapse
|
8
|
Consonni F, Gambineri E, Favre C. ALPS, FAS, and beyond: from inborn errors of immunity to acquired immunodeficiencies. Ann Hematol 2022; 101:469-484. [PMID: 35059842 PMCID: PMC8810460 DOI: 10.1007/s00277-022-04761-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/11/2022] [Indexed: 12/13/2022]
Abstract
Autoimmune lymphoproliferative syndrome (ALPS) is a primary immune regulatory disorder characterized by benign or malignant lymphoproliferation and autoimmunity. Classically, ALPS is due to mutations in FAS and other related genes; however, recent research revealed that other genes could be responsible for similar clinical features. Therefore, ALPS classification and diagnostic criteria have changed over time, and several ALPS-like disorders have been recently identified. Moreover, mutations in FAS often show an incomplete penetrance, and certain genotypes have been associated to a dominant or recessive inheritance pattern. FAS mutations may also be acquired or could become pathogenic when associated to variants in other genes, delineating a possible digenic type of inheritance. Intriguingly, variants in FAS and increased TCR αβ double-negative T cells (DNTs, a hallmark of ALPS) have been identified in multifactorial autoimmune diseases, while FAS itself could play a potential role in carcinogenesis. These findings suggest that alterations of FAS-mediated apoptosis could trespass the universe of inborn errors of immunity and that somatic mutations leading to ALPS could only be the tip of the iceberg of acquired immunodeficiencies.
Collapse
Affiliation(s)
- Filippo Consonni
- Anna Meyer Children's Hospital, University of Florence, Florence, Italy
| | - Eleonora Gambineri
- Division of Pediatric Oncology/Hematology, BMT Unit, Meyer University Children's Hospital, Viale Gaetano Pieraccini 24, 50139, Florence, Italy.
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy.
| | - Claudio Favre
- Division of Pediatric Oncology/Hematology, BMT Unit, Meyer University Children's Hospital, Viale Gaetano Pieraccini 24, 50139, Florence, Italy
| |
Collapse
|
9
|
Goode E, Smith JA, Gilpatrick M, Matney C, Borch-Christensen B, Henry MM. Autoimmune lymphoproliferative syndrome with Langerhans cell histiocytosis diagnosis. Pediatr Blood Cancer 2022; 69:e29301. [PMID: 34398530 DOI: 10.1002/pbc.29301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 11/06/2022]
Affiliation(s)
- Erin Goode
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, Arizona, USA.,Department of Child Health, University of Arizona College of Medicine, Phoenix, Arizona, USA
| | - Jaron A Smith
- Department of Pediatrics, Phoenix Children's Hospital, Phoenix, Arizona, USA
| | - Maryam Gilpatrick
- Department of Pediatrics, Phoenix Children's Hospital, Phoenix, Arizona, USA
| | - Chelsea Matney
- Department of Pediatrics, Phoenix Children's Hospital, Phoenix, Arizona, USA
| | - Bo Borch-Christensen
- Department of Child Health, University of Arizona College of Medicine, Phoenix, Arizona, USA.,Department of Hospital Medicine, Phoenix Children's Hospital, Phoenix, Arizona, USA
| | - Michael M Henry
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, Arizona, USA.,Department of Child Health, University of Arizona College of Medicine, Phoenix, Arizona, USA
| |
Collapse
|
10
|
Hafezi N, Zaki-Dizaji M, Nirouei M, Asadi G, Sharifinejad N, Jamee M, Erfan Rasouli S, Hamedifar H, Sabzevari A, Chavoshzadeh Z, Yazdani R, Abolhassani H, Aghamohammadi A, Azizi G. Clinical, immunological, and genetic features in 780 patients with autoimmune lymphoproliferative syndrome (ALPS) and ALPS-like diseases: A systematic review. Pediatr Allergy Immunol 2021; 32:1519-1532. [PMID: 33963613 DOI: 10.1111/pai.13535] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 04/23/2021] [Accepted: 04/30/2021] [Indexed: 01/07/2023]
Abstract
BACKGROUND Autoimmune lymphoproliferative syndrome (ALPS) is a group of genetic disorders characterized by early-onset lymphoproliferation, autoimmune cytopenias, and susceptibility to lymphoma. The majority of ALPS patients carry heterozygous germline mutations in the TNFRSF6 gene. In this study, we conducted a systematic review of patients with ALPS and ALPS-like syndrome. METHODS The literature search was performed in Web of Science, Scopus, and PubMed databases to find eligible studies. Additionally, the reference list of all included papers was hand-searched for additional studies. Demographic, clinical, immunological, and molecular data were extracted and compared between the ALPS and ALPS-like syndrome. RESULTS Totally, 720 patients with ALPS (532 genetically determined and 189 genetically undetermined ALPS) and 59 cases with ALPS-like phenotype due to mutations in genes other than ALPS genes were assessed. In both ALPS and ALPS-like patients, splenomegaly was the most common clinical presentation followed by autoimmune cytopenias and lymphadenopathy. Among other clinical manifestations, respiratory tract infections were significantly higher in ALPS-like patients than ALPS. The immunological analysis showed a lower serum level of IgA, IgG, and lymphocyte count in ALPS-like patients compared to ALPS. Most (85%) of the ALPS and ALPS-like cases with determined genetic defects carry mutations in the FAS gene. About one-third of patients received immunosuppressive therapy with conventional or targeted immunotherapy agents. A small fraction of patients (3.3%) received hematopoietic stem cell transplantation with successful engraftment, and all except two patients survived after transplantation. CONCLUSION Our results showed that the FAS gene with 85% frequency is the main etiological cause of genetically diagnosed patients with ALPS phenotype; therefore, the genetic defect of the majority of suspected ALPS patients could be confirmed by mutation analysis of FAS gene.
Collapse
Affiliation(s)
- Nasim Hafezi
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Majid Zaki-Dizaji
- Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Matineh Nirouei
- Student Research Committee, Alborz University of Medical Sciences, Karaj, Iran.,Alborz Office of USERN, Universal Scientific Education and Research Network (USERN, Alborz University of Medical Sciences, Karaj, Iran
| | - Gelayol Asadi
- Student Research Committee, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Niusha Sharifinejad
- Student Research Committee, Alborz University of Medical Sciences, Karaj, Iran.,Alborz Office of USERN, Universal Scientific Education and Research Network (USERN, Alborz University of Medical Sciences, Karaj, Iran
| | - Mahnaz Jamee
- Pediatric Infections Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Seyed Erfan Rasouli
- Student Research Committee, Alborz University of Medical Sciences, Karaj, Iran.,Alborz Office of USERN, Universal Scientific Education and Research Network (USERN, Alborz University of Medical Sciences, Karaj, Iran
| | - Haleh Hamedifar
- CinnaGen Medical Biotechnology Research Center, Alborz University of medical sciences, Karaj, Iran.,CinnaGen Research and production Co, Alborz, Iran
| | - Araz Sabzevari
- CinnaGen Medical Biotechnology Research Center, Alborz University of medical sciences, Karaj, Iran.,Orchid pharmed company, Tehran, Iran
| | - Zahra Chavoshzadeh
- Pediatric Infections Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Immunology and Allergy Department, Mofid Children's Hospital, Shahid Beheshti University of Medical Sciences, Tehran, 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
| | - Gholamreza Azizi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Matas Pérez E, Valdivieso Shephard JL, Bravo García-Morato M, Robles Marhuenda Á, Martinez-Ojinaga Nodal E, Prieto Bozano G, González Casado I, Salamanca Fresno L, Méndez Echevarria A, Del Rosal Rabes T, Allende Martínez L, López-Granados E, Rodríguez Pena R. Variants in CASP10, a diagnostic challenge: Single center experience and review of the literature. Clin Immunol 2021; 230:108812. [PMID: 34329798 DOI: 10.1016/j.clim.2021.108812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/21/2021] [Accepted: 07/25/2021] [Indexed: 11/27/2022]
Abstract
Autoimmune lymphoproliferative syndrome is a primary immunodeficiency caused by variants in FAS-mediated apoptosis related genes and is characterized by lymphadenopathy, splenomegaly and autoimmunity. A total of six different variants in CASP10 have been described as potential causative of disease, although two of them have recently been considered polymorphisms. The high allele frequency of these variants in healthy population in addition to the broad clinical spectrum of the disease difficult the interpretation of their pathogenicity. Here, we describe the clinical and analytical findings of three new patients carrying variants in CASP10 and summarize 12 more cases from the literature. Autoimmune cytopenias, adenopathies and increment of TCRαβ+CD4-CD8- cells have been the most common findings, being possibly the FAS-mediated apoptosis pathway the pathogenic mechanism of this disease. The clinical impact and the consequences of CASP10 variants are not fully elucidated, therefore the description of new cases will contribute to solve this issue.
Collapse
Affiliation(s)
| | | | - María Bravo García-Morato
- Department of Immunology, La Paz University Hospital, Madrid, Spain; Center for Biomedical Network Research on Rare Diseases (CIBERER U767), Madrid, Spain; La Paz Institute of Biomedical Research, Madrid, Spain.
| | - Ángel Robles Marhuenda
- Center for Biomedical Network Research on Rare Diseases (CIBERER U767), Madrid, Spain; La Paz Institute of Biomedical Research, Madrid, Spain; Department of Internal Medicine, La Paz University Hospital, Madrid, Spain.
| | | | - Gerardo Prieto Bozano
- Department of Pediatric Gastroenterology, La Paz University Hospital, Madrid, Spain.
| | | | | | - Ana Méndez Echevarria
- Department of Pediatric Infectious Diseases, La Paz University Hospital, Madrid, Spain.
| | | | - Luis Allende Martínez
- Department of Immunology, 12 de Octubre University Hospital, Madrid, Spain; Research Institute Hospital 12 Octubre (I+12), Madrid, Spain.
| | - Eduardo López-Granados
- Department of Immunology, La Paz University Hospital, Madrid, Spain; Center for Biomedical Network Research on Rare Diseases (CIBERER U767), Madrid, Spain; La Paz Institute of Biomedical Research, Madrid, Spain.
| | - Rebeca Rodríguez Pena
- Department of Immunology, La Paz University Hospital, Madrid, Spain; Center for Biomedical Network Research on Rare Diseases (CIBERER U767), Madrid, Spain; La Paz Institute of Biomedical Research, Madrid, Spain.
| |
Collapse
|
12
|
Magerus A, Bercher-Brayer C, Rieux-Laucat F. The genetic landscape of the FAS pathway deficiencies. Biomed J 2021; 44:388-399. [PMID: 34171534 PMCID: PMC8514852 DOI: 10.1016/j.bj.2021.06.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/04/2021] [Accepted: 06/17/2021] [Indexed: 02/06/2023] Open
Abstract
Dysfunction of the FAS-FASLG pathway causes a lymphoproliferative disorder with autoimmunity called Autoimmune lymphoproliferative syndrome (ALPS) mainly caused by FAS mutations. The goal of this review is to describe the genetic bases of the autoimmune lymphoproliferative syndrome and to underline their genetic complexity with the contribution of both germline and somatic events accounting for the variable clinical penetrance of the FAS mutations. Starting from the cohort of patients studied in the French cohort (>165 cases), we also reviewed the literature cases in order to depict a full description of the mutations affecting the FAS-FASLG pathway involved in the outcome of this rare non-malignant and non-infectious pediatric lymphoproliferative disease. We also discussed the variable clinical penetrance associated with mutations affecting the extracellular domain of the protein. Such non-penetrant germline mutations are frequently associated with an additional somatic event impacting the second allele of FAS. Moreover, the uncomplete clinical penetrance associated with mutations affecting the intracellular domain of FAS, in patient lacking additional FAS somatic event, suggested a potential digenic inheritance with a FAS mutation accompanied by a genetic modifier possibly impacting another player of the lymphocytes homeostasis (affecting the survival, activation or apoptosis of the peripheral leukocytes).
Collapse
Affiliation(s)
- Aude Magerus
- University of Paris, Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, INSERM UMR 1163, Paris, France.
| | - Clara Bercher-Brayer
- University of Paris, Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Frédéric Rieux-Laucat
- University of Paris, Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, INSERM UMR 1163, Paris, France
| |
Collapse
|
13
|
Casamayor-Polo L, López-Nevado M, Paz-Artal E, Anel A, Rieux-Laucat F, Allende LM. Immunologic evaluation and genetic defects of apoptosis in patients with autoimmune lymphoproliferative syndrome (ALPS). Crit Rev Clin Lab Sci 2020; 58:253-274. [PMID: 33356695 DOI: 10.1080/10408363.2020.1855623] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Apoptosis plays an important role in controlling the adaptive immune response and general homeostasis of the immune cells, and impaired apoptosis in the immune system results in autoimmunity and immune dysregulation. In the last 25 years, inherited human diseases of the Fas-FasL pathway have been recognized. Autoimmune lymphoproliferative syndrome (ALPS) is an inborn error of immunity, characterized clinically by nonmalignant and noninfectious lymphoproliferation, autoimmunity, and increased risk of lymphoma due to a defect in lymphocyte apoptosis. The laboratory hallmarks of ALPS are an elevated percentage of T-cell receptor αβ double negative T cells (DNTs), elevated levels of vitamin B12, soluble FasL, IL-10, IL-18 and IgG, and defective in vitro Fas-mediated apoptosis. In order of frequency, the genetic defects associated with ALPS are germinal and somatic ALPS-FAS, ALPS-FASLG, ALPS-CASP10, ALPS-FADD, and ALPS-CASP8. Partial disease penetrance and severity suggest the combination of germline and somatic FAS mutations as well as other risk factor genes. In this report, we summarize human defects of apoptosis leading to ALPS and defects that are known as ALPS-like syndromes that can be clinically similar to, but are genetically distinct from, ALPS. An efficient genetic and immunological diagnostic approach to patients suspected of having ALPS or ALPS-like syndromes is essential because this enables the establishment of specific therapeutic strategies for improving the prognosis and quality of life of patients.
Collapse
Affiliation(s)
- Laura Casamayor-Polo
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Marta López-Nevado
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Estela Paz-Artal
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain.,Immunology Department, University Hospital 12 de Octubre, Madrid, Spain.,School of Medicine, University Hospital 12 de Octubre, Complutense University of Madrid, Madrid, Spain
| | - Alberto Anel
- Apoptosis, Immunity and Cancer Group, University of Zaragoza/Aragón Health Research Institute (IIS-Aragón), Zaragoza, Spain
| | - Frederic Rieux-Laucat
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Université de Paris, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Luis M Allende
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain.,Immunology Department, University Hospital 12 de Octubre, Madrid, Spain.,School of Medicine, University Hospital 12 de Octubre, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
14
|
Key diagnostic markers for autoimmune lymphoproliferative syndrome with molecular genetic diagnosis. Blood 2020; 136:1933-1945. [DOI: 10.1182/blood.2020005486] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/29/2020] [Indexed: 01/01/2023] Open
Abstract
Abstract
Autoimmune lymphoproliferative syndrome (ALPS) is a rare immunodeficiency caused by mutations in genes affecting the extrinsic apoptotic pathway (FAS, FASL, CASP10). This study evaluated the clinical manifestations, laboratory findings, and molecular genetic results of 215 patients referred as possibly having ALPS. Double-negative T-cell (DNT) percentage and in vitro apoptosis functional tests were evaluated by fluorescence-activated cell sorting; interleukin 10 (IL-10) and IL-18 and soluble FAS ligand (sFASL) were measured by enzyme-linked immunosorbent assay. Genetic analysis was performed by next-generation sequencing. Clinical background data were collected from patients’ records. Patients were categorized into definite, suspected, or unlikely ALPS groups, and laboratory parameters were compared among these groups. Of 215 patients, 38 met the criteria for definite ALPS and 17 for suspected ALPS. The definite and suspected ALPS patient populations showed higher DNT percentages than unlikely ALPS and had higher rates of lymphoproliferation. Definite ALPS patients had a significantly more abnormal in vitro apoptosis function, with lower annexin, than patients with suspected ALPS (P = .002) and patients not meeting ALPS criteria (P < .001). The combination of elevated DNTs and an abnormal in vitro apoptosis functional test was the most useful in identifying all types of ALPS patients; the combination of an abnormal in vitro apoptosis functional test and elevated sFASLs was a predictive marker for ALPS-FAS group identification. Lymphoproliferation, apoptosis functional test, and DNTs are the most sensitive markers; elevated IL-10 and IL-18 are additional indicators for ALPS. The combination of elevated sFASLs and abnormal apoptosis function was the most valuable prognosticator for patients with FAS mutations.
Collapse
|
15
|
Thrombotic thrombocytopenic purpura and defective apoptosis due to CASP8/10 mutations: the role of mycophenolate mofetil. Blood Adv 2020; 3:3432-3435. [PMID: 31714957 DOI: 10.1182/bloodadvances.2019000575] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/17/2019] [Indexed: 01/19/2023] Open
Abstract
Key Points
Immunological dysregulation may underlie unusual autoimmune diseases, which also deserve to be investigated from a genetic point of view.
Collapse
|
16
|
Speir M, Lawlor KE. RIP-roaring inflammation: RIPK1 and RIPK3 driven NLRP3 inflammasome activation and autoinflammatory disease. Semin Cell Dev Biol 2020; 109:114-124. [PMID: 32771377 DOI: 10.1016/j.semcdb.2020.07.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/13/2020] [Accepted: 07/16/2020] [Indexed: 01/05/2023]
Abstract
Autoinflammatory syndromes comprise a spectrum of clinical disorders characterised by recurrent, inflammatory episodes, many of which result from the release of the pro-inflammatory cytokine, interleukin-1β (IL-1β). Inflammation and programmed cell death are tightly linked, and lytic forms of cell death, such as necroptosis and pyroptosis, are considered to be inflammatory due to the release of damage-associated molecular patterns (DAMPs). In contrast, apoptosis is traditionally regarded as immunologically silent. Recent studies, however, have uncovered a high degree of crosstalk between cell death and inflammatory signalling pathways, and effectively consolidated them into one interconnected network that converges on NLRP3 inflammasome-mediated activation of IL-1β. The receptor-interacting protein kinases (RIPK) 1 and 3 are central to this network, as highlighted by the fact that mutations in genes encoding repressors of RIPK1 and/or RIPK3 activity can lead to heightened inflammation, particularly via NLRP3 inflammasome activation. In this review, we give an overview of extrinsic cell death and inflammatory signalling pathways, and then highlight the growing number of autoinflammatory diseases that are associated with aberrant cell death and inflammasome activation.
Collapse
Affiliation(s)
- Mary Speir
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, Victoria, Australia; Department of Molecular and Translational Science, Monash University, Melbourne, Victoria, Australia.
| | - Kate E Lawlor
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, Victoria, Australia; Department of Molecular and Translational Science, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
17
|
Thaler FS, Bangol B, Biljecki M, Havla J, Schumacher AM, Kümpfel T. Possible link of genetic variants to autoimmunity in GAD-antibody-associated neurological disorders. J Neurol Sci 2020; 413:116860. [PMID: 32388243 DOI: 10.1016/j.jns.2020.116860] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 04/07/2020] [Accepted: 04/26/2020] [Indexed: 12/31/2022]
Abstract
OBJECTIVE In patients with GAD-antibody (ab) associated neurological disorders coexistence of other autoimmune disorders is observed. METHODS In this exploratory study we analysed variations in 33 candidate genes involved in autoimmunity or representing immunological check-points using next-generation sequencing. We performed haplotype-analysis of HLA-DRB1 and HLA-DQB1. Additionally, we analysed levels of sFasL, IL10, and IL18 in serum of patients and healthy controls. RESULTS 19 patients (3 males, 16 females; mean age at onset: 46.4 years) with positive GAD-ab and the following neurological phenotypes were included: n = 8 cerebellar ataxia, n = 6 limbic encephalitis, n = 4 stiff person syndrome, n = 1 demyelinating CNS disease with recurrent optic neuritis. 15 patients exhibited at least one other autoimmune disorder and/or showed other auto-ab. We identified several variations in genes linked to autoimmunity or representing check-point proteins. Most frequently (14/19 patients, allele frequency: 42.1%), we observed an amino acid exchange in the cytotoxic T-lymphocyte-associated protein 4 (CTLA4) gene. Two of the observed variants are known to cause alterations of protein function (Y446C in caspase-10, K750N in protein-tyrosin-phosphatase, non-receptor type 22). These latter variants were detected in two related patients (mother and daughter) who both present with GAD-ab-associated neurological disorders but with different clinical phenotypes. The rare haplotype DRB1*15:01:01 ~ DQA1*01:02:01 ~ DQB1*05:02:01 previously described in patients with GAD-ab-associated neurological disorders was not observed in any of our patients. No elevated serum levels of sFasL, IL18 or IL10 were observed in patients indicating no typical phenotype of autoimmune lymphoproliferate syndrome. CONCLUSIONS These findings suggest genetic risk factors in patients with GAD-ab-associated neurological disorders.
Collapse
Affiliation(s)
- Franziska S Thaler
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians University Munich, Munich, Germany.
| | - Barbara Bangol
- Center for Human Genetics and Laboratory Diagnostics, Martinsried, Germany
| | - Michelle Biljecki
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians University Munich, Munich, Germany
| | - Joachim Havla
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians University Munich, Munich, Germany
| | - Adrian-Minh Schumacher
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians University Munich, Munich, Germany
| | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians University Munich, Munich, Germany
| |
Collapse
|
18
|
Rodríguez-Rodríguez N, Flores-Mendoza G, Apostolidis SA, Rosetti F, Tsokos GC, Crispín JC. TCR-α/β CD4 - CD8 - double negative T cells arise from CD8 + T cells. J Leukoc Biol 2020; 108:851-857. [PMID: 32052478 DOI: 10.1002/jlb.1ab0120-548r] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 01/30/2020] [Accepted: 02/01/2020] [Indexed: 01/02/2023] Open
Abstract
The cellular origin of CD4- CD8- (double negative, DNT) TCR-α/β+ T cells remains unknown. Available evidence indicates that they may derive from CD8+ T cells, but most published data have been obtained using cells that bear an invariant transgenic T cell receptor that recognizes an Ag that is not present in normal mice. Here, we have used complementary fate mapping and adoptive transfer experiments to identify the cellular lineage of origin of DNT cells in wild-type mice with a polyclonal T cell repertoire. We show that TCR-α/β+ DNT cells can be traced back to CD8+ and CD4+ CD8+ double positive cells in the thymus. We also demonstrate that polyclonal DNT cells generated in secondary lymphoid organs proliferate upon adoptive transfer and can regain CD8 expression in lymphopenic environment. These results demonstrate the cellular origin of DNT cells and provide a conceptual framework to understand their presence in pathological circumstances.
Collapse
Affiliation(s)
- Noé Rodríguez-Rodríguez
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Current address: Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Giovanna Flores-Mendoza
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Sokratis A Apostolidis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Current address: Division of Rheumatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Florencia Rosetti
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - José C Crispín
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Mexico City, Mexico
| |
Collapse
|
19
|
Meynier S, Rieux-Laucat F. FAS and RAS related Apoptosis defects: From autoimmunity to leukemia. Immunol Rev 2019; 287:50-61. [PMID: 30565243 DOI: 10.1111/imr.12720] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 09/09/2018] [Indexed: 02/07/2023]
Abstract
The human adaptive immune system recognizes almost all the pathogens that we encounter and all the tumor antigens that may arise during our lifetime. Primary immunodeficiencies affecting lymphocyte development or function therefore lead to severe infections and tumor susceptibility. Furthermore, the fact that autoimmunity is a frequent feature of primary immunodeficiencies reveals a third function of the adaptive immune system: its self-regulation. Indeed, the generation of a broad repertoire of antigen receptors (via a unique strategy of random somatic rearrangements of gene segments in T cell and B cell receptor loci) inevitably creates receptors with specificity for self-antigens and thus leads to the presence of autoreactive lymphocytes. There are many different mechanisms for controlling the emergence or action of autoreactive lymphocytes, including clonal deletion in the primary lymphoid organs, receptor editing, anergy, suppression of effector lymphocytes by regulatory lymphocytes, and programmed cell death. Here, we review the genetic defects affecting lymphocyte apoptosis and that are associated with lymphoproliferation and autoimmunity, together with the role of somatic mutations and their potential involvement in more common autoimmune diseases.
Collapse
Affiliation(s)
- Sonia Meynier
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, Paris, France.,Imagine Institute, Paris Descartes-Sorbonne Paris Cité University, Paris, France
| | - Frédéric Rieux-Laucat
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, Paris, France.,Imagine Institute, Paris Descartes-Sorbonne Paris Cité University, Paris, France
| |
Collapse
|
20
|
Miano M, Cappelli E, Pezzulla A, Venè R, Grossi A, Terranova P, Palmisani E, Maggiore R, Guardo D, Lanza T, Calvillo M, Micalizzi C, Pierri F, Vernarecci C, Beccaria A, Corsolini F, Lanciotti M, Russo G, Ceccherini I, Dufour C, Fioredda F. FAS‐mediated apoptosis impairment in patients with ALPS/ALPS‐like phenotype carrying variants on
CASP10
gene. Br J Haematol 2019; 187:502-508. [DOI: 10.1111/bjh.16098] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 05/21/2019] [Indexed: 02/05/2023]
Affiliation(s)
- Maurizio Miano
- Haematology Unit IRCCS Istituto Giannina Gaslini GenoaItaly
| | | | - Agnese Pezzulla
- Haematology Unit IRCCS Istituto Giannina Gaslini GenoaItaly
- Pediatric Hematology/Oncology Unit University of Catania CataniaItaly
| | - Roberta Venè
- Molecular Oncology and Angiogenesis Unit IRCCS Ospedale Policlinico San Martino GenoaItaly
| | - Alice Grossi
- Genetic Unit IRCCS Istituto Giannina Gaslini GenoaItaly
| | | | | | | | - Daniela Guardo
- Haematology Unit IRCCS Istituto Giannina Gaslini GenoaItaly
- Haematology Clinic, Department of Internal Medicine (DiMI) University of Genoa, IRCCS AOU S. Martino‐IST GenoaItaly
| | - Tiziana Lanza
- Haematology Unit IRCCS Istituto Giannina Gaslini GenoaItaly
| | | | | | | | | | | | - Fabio Corsolini
- Laboratory of Molecular Genetics and Biobanks IRCCS Istituto Giannina Gaslini Genoa Italy
| | | | - Giovanna Russo
- Pediatric Hematology/Oncology Unit University of Catania CataniaItaly
| | | | - Carlo Dufour
- Haematology Unit IRCCS Istituto Giannina Gaslini GenoaItaly
| | | |
Collapse
|
21
|
Backus KM, Cao J, Maddox SM. Opportunities and challenges for the development of covalent chemical immunomodulators. Bioorg Med Chem 2019; 27:3421-3439. [PMID: 31204229 DOI: 10.1016/j.bmc.2019.05.050] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 05/24/2019] [Accepted: 05/31/2019] [Indexed: 02/06/2023]
Abstract
Compounds that react irreversibly with cysteines have reemerged as potent and selective tools for altering protein function, serving as chemical probes and even clinically approved drugs. The exquisite sensitivity of human immune cell signaling pathways to oxidative stress indicates the likely, yet still underexploited, general utility of covalent probes for selective chemical immunomodulation. Here, we provide an overview of immunomodulatory cysteines, including identification of electrophilic compounds available to label these residues. We focus our discussion on three protein classes essential for cell signaling, which span the 'druggability' spectrum from amenable to chemical probes (kinases), somewhat druggable (proteases), to inaccessible (phosphatases). Using existing inhibitors as a guide, we identify general strategies to guide the development of covalent probes for selected undruggable classes of proteins and propose the application of such compounds to alter immune cell functions.
Collapse
Affiliation(s)
- Keriann M Backus
- Departments of Biological Chemistry and Chemistry and Biochemistry, University of California Los Angeles, USA.
| | - Jian Cao
- Departments of Biological Chemistry and Chemistry and Biochemistry, University of California Los Angeles, USA
| | - Sean M Maddox
- Departments of Biological Chemistry and Chemistry and Biochemistry, University of California Los Angeles, USA
| |
Collapse
|
22
|
Comprehensive and Systematic Analysis of Gene Expression Patterns Associated with Body Mass Index. Sci Rep 2019; 9:7447. [PMID: 31092860 PMCID: PMC6520409 DOI: 10.1038/s41598-019-43881-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 04/29/2019] [Indexed: 12/20/2022] Open
Abstract
Both genetic and environmental factors are suggested to influence overweight and obesity risks. Although individual loci and genes have been frequently shown to be associated with body mass index (BMI), the overall interaction of these genes and their role in BMI remains underexplored. Data were collected in 90 healthy, predominately Caucasian participants (51% female) with a mean age of 26.00 ± 9.02 years. Whole blood samples were assayed by Affymetrix GeneChip Human Genome U133 Plus 2.0 Array. We integrated and analyzed the clinical and microarray gene expression data from those individuals to understand various systematic gene expression patterns underlying BMI. Conventional differential expression analysis identified seven genes RBM20, SEPT12, AX748233, SLC30A3, WTIP, CASP10, and OR12D3 associated with BMI. Weight gene co-expression network analysis among 4,647 expressed genes identified two gene modules associated with BMI. These two modules, with different extents of gene connectivity, are enriched for catabolic and muscle system processes respectively, and tend to be regulated by zinc finger transcription factors. A total of 246 hub genes were converted to non-hub genes, and 286 non-hub genes were converted to hub genes between normal and overweight individuals, revealing the network dynamics underlying BMI. A total of 28 three-way gene interactions were identified, suggesting the existence of high-order gene expression patterns underlying BMI. Our study demonstrated a variety of systematic gene expression patterns associated with BMI and thus provided novel understanding regarding the genetic factors for overweight and obesity risks on system levels.
Collapse
|
23
|
The Autoimmune Lymphoproliferative Syndrome with Defective FAS or FAS-Ligand Functions. J Clin Immunol 2018; 38:558-568. [PMID: 29911256 DOI: 10.1007/s10875-018-0523-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 06/06/2018] [Indexed: 02/08/2023]
Abstract
The autoimmune lymphoproliferative syndrome (ALPS) is a non-malignant and non-infectious uncontrolled proliferation of lymphocytes accompanied by autoimmune cytopenia. The genetic etiology of the ALPS was described in 1995 by the discovery of the FAS gene mutations. The related apoptosis defect accounts for the accumulation of autoreactive lymphocytes as well as for specific clinical and biological features that distinguish the ALPS-FAS from other monogenic defects of this apoptosis pathway, such as FADD and CASPASE 8 deficiencies. The ALPS-FAS was the first description of a monogenic cause of autoimmunity, but its non-Mendelian expression remained elusive until the description of somatic and germline mutations in ALPS patients. The recognition of these genetic diseases brought new information on the role of this apoptotic pathway in controlling the adaptive immune response in humans.
Collapse
|
24
|
Zheng L, Li J, Lenardo M. Restimulation-induced cell death: new medical and research perspectives. Immunol Rev 2018; 277:44-60. [PMID: 28462523 DOI: 10.1111/imr.12535] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In the periphery, homeostasis of the immune system depends on the equilibrium of expanding and contracting T lymphocytes during immune response. An important mechanism of lymphocyte contraction is clonal depletion of activated T cells by cytokine withdrawal induced death (CWID) and TCR restimulation induced cell death (RICD). Deficiencies in signaling components for CWID and RICD leads to autoimmunune lymphoproliferative disorders in mouse and human. The most important feature of CWID and RICD is clonal specificity, which lends great appeal as a strategy for targeted tolerance induction and treatment of autoimmune diseases, allergic disorders, and graft rejection by depleting undesired disease-causing T cells while keeping the overall host immunity intact.
Collapse
Affiliation(s)
- Lixin Zheng
- Laboratory of Immunology and Clinical Genomics Program, Molecular Development of the Immune System Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jian Li
- Laboratory of Immunology and Clinical Genomics Program, Molecular Development of the Immune System Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.,Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Michael Lenardo
- Laboratory of Immunology and Clinical Genomics Program, Molecular Development of the Immune System Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
25
|
Ben-Mustapha I, Agrebi N, Barbouche MR. Novel insights into FAS defects underlying autoimmune lymphoproliferative syndrome revealed by studies in consanguineous patients. J Leukoc Biol 2017; 103:501-508. [PMID: 29345341 DOI: 10.1002/jlb.5mr0817-332r] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 09/27/2017] [Accepted: 10/10/2017] [Indexed: 11/08/2022] Open
Abstract
Autoimmune lymphoproliferative syndrome (ALPS) is a primary immunodeficiency disease due to impaired Fas-Fas ligand apoptotic pathway. It is characterized by chronic nonmalignant, noninfectious lymphadenopathy and/or splenomegaly associated with autoimmune manifestations primarily directed against blood cells. Herein, we review the heterogeneous ALPS molecular bases and discuss recent findings revealed by the study of consanguineous patients. Indeed, this peculiar genetic background favored the identification of a novel form of AR ALPS-FAS associated with normal or residual protein expression, expanding the spectrum of ALPS types. In addition, rare mutational mechanisms underlying the splicing defects of FAS exon 6 have been identified in AR ALPS-FAS with lack of protein expression. These findings will help decipher critical regions required for the tight regulation of FAS exon 6 splicing. We also discuss the genotype-phenotype correlation and disease severity in AR ALPS-FAS. Altogether, the study of ALPS molecular bases in endogamous populations helps to better classify the disease subgroups and to unravel the Fas pathway functioning.
Collapse
Affiliation(s)
- Imen Ben-Mustapha
- Department of Immunology and LR11IPT02, Institut Pasteur de Tunis, 1002, Tunis-Belvédère, Tunisia.,The University of Tunis El Manar, Tunis, Tunisia
| | - Nourhen Agrebi
- Department of Immunology and LR11IPT02, Institut Pasteur de Tunis, 1002, Tunis-Belvédère, Tunisia.,The University of Tunis El Manar, Tunis, Tunisia.,Faculty of Sciences of Bizerte, The University of Carthage, Bizerte, Tunisia
| | - Mohamed-Ridha Barbouche
- Department of Immunology and LR11IPT02, Institut Pasteur de Tunis, 1002, Tunis-Belvédère, Tunisia.,The University of Tunis El Manar, Tunis, Tunisia
| |
Collapse
|
26
|
Khabazi A, Maralani M, Andalib S, Sakhinia E. A novel TNFRSF1A gene mutation in a patient with tumor necrosis factor receptor-associated periodic syndrome. Hematol Oncol Stem Cell Ther 2016; 11:175-177. [PMID: 27793577 DOI: 10.1016/j.hemonc.2016.09.001] [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: 07/23/2016] [Accepted: 09/26/2016] [Indexed: 11/19/2022] Open
Abstract
Tumor necrosis factor receptor-associated periodic syndrome (TRAPS) is a periodic fever syndrome inherited in an autosomal dominant fashion. It stems from mutations in the TNFRSF1A (accession number: NM_001065) gene expressing the receptor for tumor necrosis factor α. A patient with TRAPS may present with prolonged episodes of fever attacks, abdominal pain, severe myalgia, and painful erythema on the trunk or extremities. Here, we report an 8-year-old boy with febrile attacks occurring every 1-2months and continuing for 3-4days. The patient experienced 40°C-fever attacks without chills. Approximately 80% of fever attacks were accompanied by abdominal manifestations. Direct sequencing analysis was used to assess the genomic DNA of the patient, and a heterozygous R426L mutation in exon 10 of the TNFRSF1A gene in an autosomal dominant inheritance fashion was identified. Further genetic analyses were also carried out on his parents. Due to the fact that the mutation was not inherited from the parents, it was likely that R426L was a de novo and novel mutation in the TNFRSF1A gene, which can trigger TRAPS or TRAPS-like symptoms.
Collapse
Affiliation(s)
- Alireza Khabazi
- Connective Tissue Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahafarin Maralani
- Department of Molecular Medicine, Health Science Institute, Dokuz Eylul University, Izmir, Turkey
| | - Sasan Andalib
- Neuroscience Research Center, Guilan University of Medical Sciences, Rasht, Iran; Department of Neurosurgery, Poursina Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ebrahim Sakhinia
- Connective Tissue Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Molecular Medicine, Health Science Institute, Dokuz Eylul University, Izmir, Turkey; Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
27
|
Xie Y, Pittaluga S, Price S, Raffeld M, Hahn J, Jaffe ES, Rao VK, Maric I. Bone marrow findings in autoimmune lymphoproliferative syndrome with germline FAS mutation. Haematologica 2016; 102:364-372. [PMID: 27846610 DOI: 10.3324/haematol.2015.138081] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 10/13/2016] [Indexed: 11/09/2022] Open
Abstract
Autoimmune lymphoproliferative syndrome is a rare genetic disorder characterized by defective FAS-mediated apoptosis, autoimmune disease, accumulation of mature T-cell receptor alpha/beta positive, CD4 and CD8 double-negative T cells and increased risk of lymphoma. Despite frequent hematologic abnormalities, literature is scarce regarding the bone marrow pathology in autoimmune lymphoproliferative syndrome. We retrospectively reviewed 3l bone marrow biopsies from a cohort of 240 patients with germline FAS mutations. All biopsies were performed for the evaluation of cytopenias or to rule out lymphoma. Clinical information was collected and morphological, immunohistochemical, flow cytometric and molecular studies were performed. Bone marrow lymphocytosis was the predominant feature, present in 74% (23/31) of biopsies. The lymphoid cells showed several different patterns of infiltration, most often forming aggregates comprising T cells in 15 cases, B cells in one and a mixture of T and B cells in the other seven cases. Double-negative T cells were detected by immunohistochemistry in the minority of cases (10/31; 32%); significantly, all but one of these cases had prominent double-negative T-lymphoid aggregates, which in four cases diffusely replaced the marrow space. One case showed features of Rosai-Dorfman disease, containing scattered S-100+ cells with emperipolesis and double-negative T cells. No clonal B or T cells were detected by polymerase chain reaction in any evaluated cases. Classical Hodgkin lymphoma was identified in three cases. Our results demonstrate that infiltrates of T cells, or rarely B cells, can be extensive in patients with autoimmune lymphoproliferative syndrome, mimicking lymphoma. A multi-modality approach, integrating clinical, histological, immunohistochemical as well as other ancillary tests, can help avoid this diagnostic pitfall. This study is registered at Clinicaltrials.gov ID # NCT00001350.
Collapse
Affiliation(s)
- Yi Xie
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Stefania Pittaluga
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Susan Price
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mark Raffeld
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jamie Hahn
- Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Elaine S Jaffe
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - V Koneti Rao
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Irina Maric
- Clinical Center, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
28
|
Atypical presentation of autoimmune lymphoproliferative syndrome due to CASP10 mutation. Immunol Lett 2016; 177:22-4. [DOI: 10.1016/j.imlet.2016.07.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 06/29/2016] [Accepted: 07/01/2016] [Indexed: 01/05/2023]
|
29
|
Autoimmune lymphoproliferative syndrome due to somatic FAS mutation (ALPS-sFAS) combined with a germline caspase-10 (CASP10) variation. Immunobiology 2016; 221:40-7. [DOI: 10.1016/j.imbio.2015.08.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 07/31/2015] [Accepted: 08/11/2015] [Indexed: 12/25/2022]
|
30
|
Shah S, Wu E, Rao VK, Tarrant TK. Autoimmune lymphoproliferative syndrome: an update and review of the literature. Curr Allergy Asthma Rep 2014; 14:462. [PMID: 25086580 DOI: 10.1007/s11882-014-0462-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Autoimmune lymphoproliferative syndrome (ALPS) is characterized by immune dysregulation due to a defect in lymphocyte apoptosis. The clinical manifestations may be noted in multiple family members and include lymphadenopathy, splenomegaly, increased risk of lymphoma, and autoimmune disease, which typically involves hematopoietic cell lines manifesting as multilineage cytopenias. Since the disease was first characterized in the early 1990s, there have been many advances in the diagnosis and management of this syndrome. The inherited genetic defect of many ALPS patients has involved (FAS) pathway signaling proteins, but there remain those patients who carry undefined genetic defects. Despite ALPS having historically been considered a primary immune defect presenting in early childhood, adult onset presentation is increasingly becoming recognized and more so in genetically undefined patients and those with somatic FAS mutations. Thus, future research may identify novel pathways and/or regulatory proteins important in lymphocyte activation and apoptosis.
Collapse
Affiliation(s)
- Shaili Shah
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA,
| | | | | | | |
Collapse
|
31
|
Gerola S, Nittka S, Kähler G, Tao S, Brenner H, Binelli G, Eils R, Brors B, Neumaier M. Genetic variants in apoptosis-related genes associated with colorectal hyperplasia. Genes Chromosomes Cancer 2014; 53:769-78. [DOI: 10.1002/gcc.22185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Accepted: 04/21/2014] [Indexed: 12/13/2022] Open
Affiliation(s)
- Stefano Gerola
- Institute for Clinical Chemistry; Universitätsmedizin Mannheim, Medical Faculty Mannheim, University of Heidelberg; Mannheim 68167 Germany
| | - Stefanie Nittka
- Institute for Clinical Chemistry; Universitätsmedizin Mannheim, Medical Faculty Mannheim, University of Heidelberg; Mannheim 68167 Germany
| | - Georg Kähler
- Department of Medical; Medical Center Mannheim, Universitätsmedizin Mannheim, University of Heidelberg; Mannheim Germany
| | - Sha Tao
- Division of Clinical Epidemiology and Aging Research; German Cancer Research Center (DKFZ); Heidelberg 69120 Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research; German Cancer Research Center (DKFZ); Heidelberg 69120 Germany
| | - Giorgio Binelli
- Department of Theoretical and Applied Sciences; Insubria University; Varese Italy
| | - Roland Eils
- Division of Theoretical Bioinformatics; German Cancer Research Center (DKFZ), Im Neuenheimer Feld 224; Heidelberg Germany
| | - Benedikt Brors
- Division of Theoretical Bioinformatics; German Cancer Research Center (DKFZ), Im Neuenheimer Feld 224; Heidelberg Germany
| | - Michael Neumaier
- Institute for Clinical Chemistry; Universitätsmedizin Mannheim, Medical Faculty Mannheim, University of Heidelberg; Mannheim 68167 Germany
| |
Collapse
|
32
|
Price S, Shaw PA, Seitz A, Joshi G, Davis J, Niemela JE, Perkins K, Hornung RL, Folio L, Rosenberg PS, Puck JM, Hsu AP, Lo B, Pittaluga S, Jaffe ES, Fleisher TA, Rao VK, Lenardo MJ. Natural history of autoimmune lymphoproliferative syndrome associated with FAS gene mutations. Blood 2014; 123:1989-99. [PMID: 24398331 PMCID: PMC3968385 DOI: 10.1182/blood-2013-10-535393] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 12/31/2013] [Indexed: 12/30/2022] Open
Abstract
Autoimmune lymphoproliferative syndrome (ALPS) presents in childhood with nonmalignant lymphadenopathy and splenomegaly associated with a characteristic expansion of mature CD4 and CD8 negative or double negative T-cell receptor αβ(+) T lymphocytes. Patients often present with chronic multilineage cytopenias due to autoimmune peripheral destruction and/or splenic sequestration of blood cells and have an increased risk of B-cell lymphoma. Deleterious heterozygous mutations in the FAS gene are the most common cause of this condition, which is termed ALPS-FAS. We report the natural history and pathophysiology of 150 ALPS-FAS patients and 63 healthy mutation-positive relatives evaluated in our institution over the last 2 decades. Our principal findings are that FAS mutations have a clinical penetrance of <60%, elevated serum vitamin B12 is a reliable and accurate biomarker of ALPS-FAS, and the major causes of morbidity and mortality in these patients are the overwhelming postsplenectomy sepsis and development of lymphoma. With longer follow-up, we observed a significantly greater relative risk of lymphoma than previously reported. Avoiding splenectomy while controlling hypersplenism by using corticosteroid-sparing treatments improves the outcome in ALPS-FAS patients. This trial was registered at www.clinicaltrials.gov as #NCT00001350.
Collapse
Affiliation(s)
- Susan Price
- Molecular Development Section, Laboratory of Immunology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW Several autoimmune lymphoproliferative syndromes have been described lately. We review here the main clinical and laboratory findings of these new disorders. RECENT FINDINGS The prototypical autoimmune lymphoproliferative syndrome (ALPS) has had its diagnostic criteria modified, somatic mutations in RAS genes were found to cause an ALPS-like syndrome in humans, and mutations in a gene encoding a protein kinase C (PRKCD) were discovered to cause a syndrome of lymphoproliferation, autoimmunity and natural killer cell defect. SUMMARY The recent discoveries shed light on the molecular pathways governing lymphocyte death, proliferation and immune tolerance in humans.
Collapse
|
34
|
Su HC, Snow AL, Lenardo MJ. Programmed cell death in lymphocytes and associated disorders. Clin Immunol 2013. [DOI: 10.1016/b978-0-7234-3691-1.00037-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
35
|
Ben-Chetrit E, Bnaya A, Vernea F, Somech R, Ashkenazi YJ. Sometimes double negative is positive. Arthritis Care Res (Hoboken) 2012; 65:161-8. [PMID: 22933320 DOI: 10.1002/acr.21832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 08/13/2012] [Indexed: 11/10/2022]
Affiliation(s)
- Eli Ben-Chetrit
- Shaare Zedek Medical Center, Hebrew University School of Medicine, Jerusalem, Israel.
| | | | | | | | | |
Collapse
|
36
|
Abstract
Autoimmune lymphoproliferative syndrome (ALPS) represents a failure of apoptotic mechanisms to maintain lymphocyte homeostasis, permitting accumulation of lymphoid mass and persistence of autoreactive cells that often manifest in childhood with chronic nonmalignant lymphadenopathy, hepatosplenomegaly, and recurring multilineage cytopenias. Cytopenias in these patients can be the result of splenic sequestration as well as autoimmune complications manifesting as autoimmune hemolytic anemia, immune-mediated thrombocytopenia, and autoimmune neutropenia. More than 300 families with hereditary ALPS have now been described; nearly 500 patients from these families have been studied and followed worldwide over the last 20 years by our colleagues and ourselves. Some of these patients with FAS mutations affecting the intracellular portion of the FAS protein also have an increased risk of B-cell lymphoma. The best approaches to diagnosis, follow-up, and management of ALPS, its associated cytopenias, and other complications resulting from infiltrative lymphoproliferation and autoimmunity are presented.
Collapse
|
37
|
Tadaki H, Saitsu H, Kanegane H, Miyake N, Imagawa T, Kikuchi M, Hara R, Kaneko U, Kishi T, Miyamae T, Nishimura A, Doi H, Tsurusaki Y, Sakai H, Yokota S, Matsumoto N. Exonic deletion of CASP10 in a patient presenting with systemic juvenile idiopathic arthritis, but not with autoimmune lymphoproliferative syndrome type IIa. Int J Immunogenet 2011; 38:287-93. [DOI: 10.1111/j.1744-313x.2011.01005.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
38
|
Abstract
Autoimmune disease affects a significant proportion of the population. The etiology of most autoimmune diseases is largely unknown, but it is thought to be multifactorial with both environmental and genetic influences. Rare monogenic autoimmune diseases, however, offer an invaluable window into potential disease mechanisms. In this review, we will discuss the autoimmune polyglandular syndrome (APS1), the immunedysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX), and autoimmune lymphoproliferative syndrome (ALPS). Significantly, the information gained from the study of these diseases has provided new insights into more common autoimmune disease and have yielded new diagnostics and therapeutic opportunities.
Collapse
Affiliation(s)
- Michael Waterfield
- Diabetes Center – University of California San Francisco
- Department of Pediatrics- University of California San Francisco
| | | |
Collapse
|
39
|
Lafont E, Milhas D, Teissié J, Therville N, Andrieu-Abadie N, Levade T, Benoist H, Ségui B. Caspase-10-dependent cell death in Fas/CD95 signalling is not abrogated by caspase inhibitor zVAD-fmk. PLoS One 2010; 5:e13638. [PMID: 21049020 PMCID: PMC2964310 DOI: 10.1371/journal.pone.0013638] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2010] [Accepted: 10/06/2010] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Upon CD95/Fas ligation, the initiator caspase-8 is known to activate effector caspases leading to apoptosis. In the presence of zVAD-fmk, a broad-spectrum caspase inhibitor, Fas engagement can also trigger an alternative, non-apoptotic caspase-independent form of cell death, which is initiated by RIP1. Controversy exists as to the ability of caspase-10 to mediate cell death in response to FasL (CD95L or CD178). Herein, the role of caspase-10 in FasL-induced cell death has been re-evaluated. METHODOLOGY AND PRINCIPAL FINDINGS The present study shows that FasL-induced cell death was completely impaired in caspase-8- and caspase-10-doubly deficient (I9-2e) Jurkat leukaemia T-cell lines. Over-expressing of either caspase-8 or caspase-10 in I9-2e cells triggered cell death and restored sensitivity to FasL, further arguing for a role of both initiator caspases in Fas apoptotic signalling. In the presence of zVAD-fmk, FasL triggered an alternative form of cell death similarly in wild-type (A3) and in caspase-8-deficient Jurkat cells expressing endogenous caspase-10 (clone I9-2d). Cell death initiated by Fas stimulation in the presence of zVAD-fmk was abrogated in I9-2e cells as well as in HeLa cells, which did not express endogenous caspase-10, indicating that caspase-10 somewhat participates in this alternative form of cell death. Noteworthy, ectopic expression of caspase-10 in I9-2e and HeLa cells restored the ability of FasL to trigger cell death in the presence of zVAD-fmk. As a matter of fact, FasL-triggered caspase-10 processing still occurred in the presence of zVAD-fmk. CONCLUSIONS AND SIGNIFICANCE Altogether, these data provide genetic evidence for the involvement of initiator caspase-10 in FasL-induced cell death and indicate that zVAD-fmk does not abrogate caspase-10 processing and cytotoxicity in Fas signalling. Our study also questions the existence of an alternative caspase-independent cell death pathway in Fas signalling.
Collapse
Affiliation(s)
- Elodie Lafont
- U858 INSERM (Institut National de la Santé et de la Recherche Médicale), Département Cancer, Equipe 14, Toulouse, France
- Institut Fédératif de Recherche 150, Institut de Médecine Moléculaire de Rangueil, Toulouse, France
- Université Paul Sabatier (Toulouse III), Faculté des Sciences Pharmaceutiques, Toulouse, France
| | - Delphine Milhas
- U858 INSERM (Institut National de la Santé et de la Recherche Médicale), Département Cancer, Equipe 14, Toulouse, France
- Institut Fédératif de Recherche 150, Institut de Médecine Moléculaire de Rangueil, Toulouse, France
| | - Justin Teissié
- IPBS (Institut de Pharmacologie et de Biologie Structurale) Unité Mixte de Recherche 5089 CNRS (Centre National de la Recherche Scientifique), Toulouse, France
| | - Nicole Therville
- U858 INSERM (Institut National de la Santé et de la Recherche Médicale), Département Cancer, Equipe 14, Toulouse, France
- Institut Fédératif de Recherche 150, Institut de Médecine Moléculaire de Rangueil, Toulouse, France
| | - Nathalie Andrieu-Abadie
- U858 INSERM (Institut National de la Santé et de la Recherche Médicale), Département Cancer, Equipe 14, Toulouse, France
- Institut Fédératif de Recherche 150, Institut de Médecine Moléculaire de Rangueil, Toulouse, France
| | - Thierry Levade
- U858 INSERM (Institut National de la Santé et de la Recherche Médicale), Département Cancer, Equipe 14, Toulouse, France
- Institut Fédératif de Recherche 150, Institut de Médecine Moléculaire de Rangueil, Toulouse, France
| | - Hervé Benoist
- U858 INSERM (Institut National de la Santé et de la Recherche Médicale), Département Cancer, Equipe 14, Toulouse, France
- Institut Fédératif de Recherche 150, Institut de Médecine Moléculaire de Rangueil, Toulouse, France
- Université Paul Sabatier (Toulouse III), Faculté des Sciences Pharmaceutiques, Toulouse, France
| | - Bruno Ségui
- U858 INSERM (Institut National de la Santé et de la Recherche Médicale), Département Cancer, Equipe 14, Toulouse, France
- Institut Fédératif de Recherche 150, Institut de Médecine Moléculaire de Rangueil, Toulouse, France
- Université Paul Sabatier (Toulouse III), Faculté des Sciences Pharmaceutiques, Toulouse, France
- * E-mail:
| |
Collapse
|
40
|
Abstract
Immunoglobulin E (IgE) is a key mediator of anti-parasitic and anti-tumour immunity. However it is also a critical component of atopic and autoimmune diseases, and elevated serum IgE levels are a common indicator of immune dysregulation. In this review we survey the literature on genetic associations of elevated IgE in humans and mice. We find that defects in a limited number of pathways explain the majority of gene associations with IgE. Commonly, elevated IgE is associated with defects in Th bias and B cell class switching, severe T cell tolerance defects and defects in immunity at the host-environment interface. These genetic data demonstrate the mechanisms of control over IgE production and the manner in which they can be circumvented.
Collapse
Affiliation(s)
- John Altin
- John Curtin School of Medical Research, Australian National University, Canberra 0200, Australia
| | | | | |
Collapse
|
41
|
Rao VK, Price S, Perkins K, Aldridge P, Tretler J, Davis J, Dale JK, Gill F, Hartman KR, Stork LC, Gnarra DJ, Krishnamurti L, Newburger PE, Puck J, Fleisher T. Use of rituximab for refractory cytopenias associated with autoimmune lymphoproliferative syndrome (ALPS). Pediatr Blood Cancer 2009; 52:847-52. [PMID: 19214977 PMCID: PMC2774763 DOI: 10.1002/pbc.21965] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND ALPS is a disorder of apoptosis resulting in accumulation of autoreactive lymphocytes, leading to marked lymphadenopathy, hepatosplenomegaly, and multilineage cytopenias due to splenic sequestration and/or autoimmune destruction often presenting in childhood. We summarize our experience of rituximab use during the last 8 years in 12 patients, 9 children, and 3 adults, out of 259 individuals with ALPS, belonging to 166 families currently enrolled in studies at the National Institutes of Health. METHODS Refractory immune thrombocytopenia (platelet count <20,000) in nine patients and autoimmune hemolytic anemia (AIHA) in three patients led to treatment with rituximab. Among them, seven patients had undergone prior surgical splenectomy; three had significant splenomegaly; and two had no palpable spleen. RESULTS In seven out of nine patients with ALPS and thrombocytopenia, rituximab therapy led to median response duration of 21 months (range 14-36 months). In contrast, none of the three children treated with rituximab for AIHA responded. Noted toxicities included profound and prolonged hypogammaglobulinemia in three patients requiring replacement IVIG, total absence of antibody response to polysaccharide vaccines lasting up to 4 years after rituximab infusions in one patient and prolonged neutropenia in one patient. CONCLUSION Toxicities including hypogammaglobulinemia and neutropenia constitute an additional infection risk burden, especially in asplenic individuals, and may warrant avoidance of rituximab until other immunosuppressive medication options are exhausted. Long-term follow-up of ALPS patients with cytopenias after any treatment is necessary to determine relative risks and benefits.
Collapse
Affiliation(s)
- V Koneti Rao
- Laboratory of Clinical Infectious Diseases, NIAID, and Clinical Center, National Institutes of Health, Bethesda, Maryland 20892-1888, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Galluzzi L, Joza N, Tasdemir E, Maiuri MC, Hengartner M, Abrams JM, Tavernarakis N, Penninger J, Madeo F, Kroemer G. No death without life: vital functions of apoptotic effectors. Cell Death Differ 2008; 15:1113-23. [PMID: 18309324 PMCID: PMC2917777 DOI: 10.1038/cdd.2008.28] [Citation(s) in RCA: 191] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
As a result of the genetic experiments performed in Caenorhabditis elegans, it has been tacitly assumed that the core proteins of the 'apoptotic machinery' (CED-3, -4, -9 and EGL-1) would be solely involved in cell death regulation/execution and would not exert any functions outside of the cell death realm. However, multiple studies indicate that the mammalian orthologs of these C. elegans proteins (i.e. caspases, Apaf-1 and multidomain proteins of the Bcl-2 family) participate in cell death-unrelated processes. Similarly, loss-of-function mutations of ced-4 compromise the mitotic arrest of DNA-damaged germline cells from adult nematodes, even in a context in which the apoptotic machinery is inoperative (for instance due to mutations of egl-1 or ced-3). Moreover, EGL-1 is required for the activation of autophagy in starved nematodes. Finally, the depletion of caspase-independent death effectors, such as apoptosis-inducing factor (AIF) and endonuclease G, provokes cell death-independent consequences, both in mammals and in yeast (Saccharomyces cerevisiae). These results corroborate the conjecture that any kind of protein that has previously been specifically implicated in apoptosis might have a phylogenetically conserved apoptosis-unrelated function, most likely as part of an adaptive response to cellular stress.
Collapse
Affiliation(s)
- L Galluzzi
- INSERM, U848, Villejuif, France
- Institut Gustave Roussy, Villejuif, France
- University Paris-Sud, Paris 11, Villejuif, France
| | - N Joza
- INSERM, U848, Villejuif, France
- Institut Gustave Roussy, Villejuif, France
- University Paris-Sud, Paris 11, Villejuif, France
| | - E Tasdemir
- INSERM, U848, Villejuif, France
- Institut Gustave Roussy, Villejuif, France
- University Paris-Sud, Paris 11, Villejuif, France
| | - MC Maiuri
- INSERM, U848, Villejuif, France
- Institut Gustave Roussy, Villejuif, France
- University Paris-Sud, Paris 11, Villejuif, France
- Department of Experimental Pharmacology, School of Biotechnological Sciences, University of Naples Federico II, Naples, Italy
| | - M Hengartner
- Institute of Molecular Biology, University of Zurich, Zurich, Switzerland
| | - JM Abrams
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, USA
| | - N Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion Crete, Greece
| | - J Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna, Austria and
| | - F Madeo
- Center of Molecular Biology, University of Graz, Graz, Austria
| | - G Kroemer
- INSERM, U848, Villejuif, France
- Institut Gustave Roussy, Villejuif, France
- University Paris-Sud, Paris 11, Villejuif, France
| |
Collapse
|
43
|
Abstract
Programmed cell death is important for maintaining lymphocyte homeostasis. Several human-inherited diseases with impaired apoptosis have been identified at the genetic level: autoimmune lymphoproliferative syndrome, caspase-8 deficiency state, and X-linked lymphoproliferative syndrome. These diseases feature excess lymphocyte accumulation, autoimmunity, or immunodeficiency. Elucidating their molecular pathogenesis has also provided new insights into the signaling mechanisms regulating apoptosis and lymphocyte activation.
Collapse
Affiliation(s)
- Helen C. Su
- Clinical Investigator, Human Immunological Diseases Unit, Laboratory of Host Defense, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Michael J. Lenardo
- Senior Investigator, Molecular Development Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
44
|
Autoimmune lymphoproliferative syndrome (ALPS) caused by Fas (CD95) mutation mimicking sarcoidosis. Am J Surg Pathol 2008; 32:329-34. [PMID: 18223337 DOI: 10.1097/pas.0b013e3181484f6d] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Autoimmune lymphoproliferative syndrome (ALPS) is an inherited disorder associated with defects in apoptosis, characterized by childhood onset of lymphadenopathy, splenomegaly, hyperimmunoglobulinemia, and autoimmune disease. ALPS is most frequently associated with a mutation in the cell death receptor Fas (CD95). Very rarely a mutation in caspase 10 is present. An increase of CD4/CD8 double negative T cells in the peripheral blood and lymph nodes is a feature characteristic of ALPS. Additionally, histiocytic proliferations resembling sinus histiocytosis with massive lymphadenopathy (Rosai-Dorfman disease) were reported recently in patients with ALPS. In the rare cases with a caspase 10 mutation an accumulation of dendritic cells in lymphoid organs was noted. We describe a different, sarcoidosislike, histiocytic infiltration of lymph nodes that persisted for years in a girl, that was initially supposed to suffer from sarcoidosis, but was eventually diagnosed as ALPS, associated with a missense mutation in the intracellular death domain of Fas. This sarcoidosislike histologic picture extends the spectrum of histiocytic lymph node alterations observed in ALPS and alerts of a potential diagnostic pitfall.
Collapse
|
45
|
Disorders of Apoptosis: Mechanisms for Autoimmunity in Primary Immunodeficiency Diseases. J Clin Immunol 2008; 28 Suppl 1:S20-8. [DOI: 10.1007/s10875-007-9161-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2007] [Accepted: 12/04/2007] [Indexed: 12/26/2022]
|
46
|
Programmed cell death in lymphocytes. Clin Immunol 2008. [DOI: 10.1016/b978-0-323-04404-2.10014-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
47
|
Carneiro-Sampaio M, Coutinho A. Tolerance and autoimmunity: lessons at the bedside of primary immunodeficiencies. Adv Immunol 2007; 95:51-82. [PMID: 17869610 DOI: 10.1016/s0065-2776(07)95002-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The recent progress in the genetic characterization of many primary immunodeficiencies (PIDs) allows for a better understanding of immune molecular and cellular mechanisms. The present chapter discusses associations between PIDs and autoimmune diseases (AIDs) in this new light. PIDs are classified according to the frequency of association with AIDs, defining four groups of conditions: systematic (more than 80% of all patients), strong (10-80%), mild (less than 10%), and absent (no available descriptions). Several general conclusions could be drawn: (1) pathological autoimmune (AI) manifestations are very frequently associated with PIDs, indicating that, contrary to conventional notions, antimicrobial protection and natural tolerance to body tissues share many basic mechanisms; (2) in some gene defects, association is so strong that one could speak of "monogenic" AIDs; (3) basic types of PIDs are selectively associated with AID of a particular set of target tissues; (4) while for some gene defects, current theory satisfactorily explains pathogenesis of the corresponding AID, other situations suggest extensive gaps in the present understanding of natural tolerance; and (5) not exceptionally, observations on the AI phenotype for the same gene defect in mouse and man are not concordant, perhaps owing to the limited genetic diversity of mouse models, often limited to a single mouse strain. Overall, clinical observations on PID support the new paradigm of "dominant" tolerance to self-components, in which AID owes to deficits in immune responses (i.e., in regulatory mechanisms), rather than from excessive reactivity.
Collapse
Affiliation(s)
- Magda Carneiro-Sampaio
- Department of Pediatrics, Children's Hospital, Faculdade de Medicina da Universidade de São Paulo, Brazil
| | | |
Collapse
|
48
|
Cerutti E, Campagnoli MF, Ferretti M, Garelli E, Crescenzio N, Rosolen A, Chiocchetti A, Lenardo MJ, Ramenghi U, Dianzani U. Co-inherited mutations of Fas and caspase-10 in development of the autoimmune lymphoproliferative syndrome. BMC Immunol 2007; 8:28. [PMID: 17999750 PMCID: PMC2211507 DOI: 10.1186/1471-2172-8-28] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2007] [Accepted: 11/13/2007] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Autoimmune lymphoproliferative syndrome (ALPS) is a rare inherited disorder characterized by defective function of Fas, autoimmune manifestations that predominantly involve blood cells, polyclonal accumulation of lymphocytes in the spleen and lymph nodes with lymphoadenomegaly and/or splenomegaly, and expansion of TCRalphabeta+ CD4/CD8 double-negative (DN) T cells in the peripheral blood. Most frequently, it is due to Fas gene mutations, causing ALPS type Ia (ALPS-Ia). However, other mutations, namely of the FasL gene (ALPS-Ib) and the caspase-10 gene (ALPS-II) are occasionally detected, whereas some patients do not present any known mutations (ALPS-III). Recently, mutations of the NRAS gene have been suggested to cause ALPS-IV. RESULTS This work reports two patients that are combined heterozygous for single nucleotide substitutions in the Fas and caspase-10 genes. The first patient carried a splice site defect suppressing allele expression in the Fas gene and the P501L substitution in caspase-10. The second had a mutation causing a premature stop codon (Q47X) in the Fas gene and the Y446C substitution in caspase-10. Fas expression was reduced and caspase-10 activity was decreased in both patients. In both patients, the mutations were inherited from distinct healthy parents. CONCLUSION These data strongly suggest that co-transmission of these mutation was responsible for ALPS.
Collapse
Affiliation(s)
- Elisa Cerutti
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD) and Department of Medical Science, "A. Avogadro" University of Eastern Piedmont, Novara, Italy
| | | | - Massimo Ferretti
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD) and Department of Medical Science, "A. Avogadro" University of Eastern Piedmont, Novara, Italy
| | | | | | - Angelo Rosolen
- Department of Pediatrics, University of Padua, Padua, Italy
| | - Annalisa Chiocchetti
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD) and Department of Medical Science, "A. Avogadro" University of Eastern Piedmont, Novara, Italy
| | - Michael J Lenardo
- Molecular Development Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ugo Ramenghi
- Department of Pediatrics, University of Turin, Turin, Italy
| | - Umberto Dianzani
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD) and Department of Medical Science, "A. Avogadro" University of Eastern Piedmont, Novara, Italy
| |
Collapse
|
49
|
Bi LL, Pan G, Atkinson TP, Zheng L, Dale JK, Makris C, Reddy V, McDonald JM, Siegel RM, Puck JM, Lenardo MJ, Straus SE. Dominant inhibition of Fas ligand-mediated apoptosis due to a heterozygous mutation associated with autoimmune lymphoproliferative syndrome (ALPS) Type Ib. BMC MEDICAL GENETICS 2007; 8:41. [PMID: 17605793 PMCID: PMC1931585 DOI: 10.1186/1471-2350-8-41] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2006] [Accepted: 07/02/2007] [Indexed: 01/08/2023]
Abstract
Background: Autoimmune lymphoproliferative syndrome (ALPS) is a disorder of lymphocyte homeostasis and immunological tolerance due primarily to genetic defects in Fas (CD95/APO-1; TNFRSF6), a cell surface receptor that regulates apoptosis and its signaling apparatus. Methods: Fas ligand gene mutations from ALPS patients were identified through cDNA and genomic DNA sequencing. Molecular and biochemical assessment of these mutant Fas ligand proteins were carried out by expressing the mutant FasL cDNA in mammalian cells and analysis its effects on Fas-mediated programmed cell death. Results: We found an ALPS patient that harbored a heterozygous A530G mutation in the FasL gene that replaced Arg with Gly at position 156 in the protein's extracellular Fas-binding region. This produced a dominant-interfering FasL protein that bound to the wild-type FasL protein and prevented it from effectively inducing apoptosis. Conclusion: Our data explain how a naturally occurring heterozygous human FasL mutation can dominantly interfere with normal FasL apoptotic function and lead to an ALPS phenotype, designated Type Ib.
Collapse
Affiliation(s)
- Lilia L Bi
- Center for Biologics Evaluation and Research, FDA, Rockville, Maryland, USA
| | - George Pan
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - T Prescott Atkinson
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Lixin Zheng
- Laboratory of Immunology, NIAID, NIH, Bethesda, MD 20892, USA
| | - Janet K Dale
- Laboratory of Clinical Infectious Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | - Christopher Makris
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Vishnu Reddy
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jay M McDonald
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- The Birmingham Veteran's Administration Medical Center, Birmingham, Alabama, USA
| | | | - Jennifer M Puck
- Department of Pediatrics, University of California, San Francisco, USA
| | | | - Stephen E Straus
- Laboratory of Clinical Infectious Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
50
|
Abstract
The first proapoptotic caspase, CED-3, was cloned from Caenorhabditis elegans in 1993 and shown to be essential for the developmental death of all somatic cells. Following the discovery of CED-3, caspases have been cloned from several vertebrate and invertebrate species. As reviewed in other articles in this issue of Cell Death and Differentiation, many caspases function in nonapoptotic pathways. However, as is clear from the worm studies, the evolutionarily conserved role of caspases is to execute programmed cell death. In this article, I will specifically focus on caspases that function primarily in cell death execution. In particular, the physiological function of caspases in apoptosis is discussed using examples from the worm, fly and mammals.
Collapse
Affiliation(s)
- S Kumar
- Hanson Institute, Institute of Medical and Veterinary Science, Adelaide, SA, Australia.
| |
Collapse
|