51
|
Songane M, Khair M, Saleh M. An updated view on the functions of caspases in inflammation and immunity. Semin Cell Dev Biol 2018; 82:137-149. [PMID: 29366812 DOI: 10.1016/j.semcdb.2018.01.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 12/21/2017] [Accepted: 01/02/2018] [Indexed: 12/14/2022]
Abstract
The binary classification of mammalian caspases as either apoptotic or inflammatory is now obsolete. Emerging data indicate that all mammalian caspases are intricately involved in the regulation of inflammation and immunity. They participate in embryonic and adult tissue homeostasis, control leukocyte differentiation, activation and effector functions, and mediate innate and adaptive immunity signaling. Caspases also promote host resistance by regulating anti-oxidant defense and pathogen clearance through regulation of phagosomal maturation, actin dynamics and phagosome-lysosome fusion. Beyond apoptosis, they regulate inflammatory cell death, eliciting rapid pyroptosis of infected cells, while inhibiting necroptosis-mediated tissue destruction and chronic inflammation. In this review, we describe the cellular and molecular mechanisms underlying non-apoptotic functions of caspases in inflammation and immunity and provide an updated view of their functions as central regulators of tissue homeostasis and host defense.
Collapse
Affiliation(s)
- Mario Songane
- Department of Medicine, McGill University, Montréal, Québec H3G 0B1, Canada
| | - Mostafa Khair
- Department of Medicine, McGill University, Montréal, Québec H3G 0B1, Canada
| | - Maya Saleh
- Department of Medicine, McGill University, Montréal, Québec H3G 0B1, Canada.
| |
Collapse
|
52
|
Affiliation(s)
- Dae Chul Jeong
- Division of Pediatric Rheumatology and Clinical Immunology, Department of Pediatrics, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
53
|
Muraki M. Development of expression systems for the production of recombinant human Fas ligand extracellular domain derivatives using <em>Pichia pastoris</em> and preparation of the conjugates by site-specific chemical modifications: A review. AIMS BIOENGINEERING 2018. [DOI: 10.3934/bioeng.2018.1.39] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
54
|
Rieux-Laucat F. What's up in the ALPS. Curr Opin Immunol 2017; 49:79-86. [DOI: 10.1016/j.coi.2017.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 10/02/2017] [Indexed: 10/18/2022]
|
55
|
Hashiramoto A, Konishi Y, Murayama K, Kawasaki H, Yoshida K, Tsumiyama K, Tanaka K, Mizuhara M, Shiotsuki T, Kitamura H, Komai K, Kimura T, Yagita H, Shiozawa K, Shiozawa S. A variant of death-receptor 3 associated with rheumatoid arthritis interferes with apoptosis-induction of T cell. J Biol Chem 2017; 293:1933-1943. [PMID: 29180447 PMCID: PMC5808757 DOI: 10.1074/jbc.m117.798884] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 11/14/2017] [Indexed: 12/12/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic polyarthritis of unknown etiology. To unravel the molecular mechanisms in RA, we performed targeted DNA sequencing analysis of patients with RA. This analysis identified a variant of the death receptor 3 (DR3) gene, a member of the family of apoptosis-inducing Fas genes, which contains four single-nucleotide polymorphisms (SNPs) and a 14-nucleotide deletion within exon 5 and intron 5. We found that the deletion causes the binding of splicing regulatory proteins to DR3 pre-mRNA intron 5, resulting in a portion of intron 5 becoming part of the coding sequence, thereby generating a premature stop codon. We also found that this truncated DR3 protein product lacks the death domain and forms a heterotrimer complex with wildtype DR3 that dominant-negatively inhibits ligand-induced apoptosis in lymphocytes. Myelocytes from transgenic mice expressing the human DR3 variant produced soluble truncated DR3, forming a complex with TNF-like ligand 1A (TL1A), which inhibited apoptosis induction. In summary, our results reveal that a DR3 splice variant that interferes with ligand-induced T cell responses and apoptosis may contribute to RA pathogenesis.
Collapse
Affiliation(s)
- Akira Hashiramoto
- From the Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142
| | - Yoshitake Konishi
- From the Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142
| | - Koichi Murayama
- From the Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142
| | - Hiroki Kawasaki
- From the Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142
| | - Kohsuke Yoshida
- From the Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142
| | - Ken Tsumiyama
- the Department of Medicine, Rheumatic Diseases Unit, Kyushu University Beppu Hospital, Beppu 874-0838
| | - Kimie Tanaka
- From the Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142
| | - Masaru Mizuhara
- From the Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142
| | - Toshio Shiotsuki
- From the Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142
| | - Hitomi Kitamura
- From the Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142
| | - Koichiro Komai
- From the Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142
| | - Tomoatsu Kimura
- the Department of Orthopedic Surgery, Faculty of Medicine, University of Toyama, 3190 Gofuku, 930-0194 Toyama
| | - Hideo Yagita
- the Department of Immunology, Juntendo University School of Medicine, Tokyo 113-8431, and
| | - Kazuko Shiozawa
- the Department of Rheumatology, Hyogo Prefectural Kakogawa Medical Center, Kakogawa 675-8555, Japan
| | - Shunichi Shiozawa
- the Department of Medicine, Rheumatic Diseases Unit, Kyushu University Beppu Hospital, Beppu 874-0838,
| |
Collapse
|
56
|
Costa-Reis P, Sullivan KE. Monogenic lupus: it's all new! Curr Opin Immunol 2017; 49:87-95. [PMID: 29100097 DOI: 10.1016/j.coi.2017.10.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 10/10/2017] [Indexed: 02/01/2023]
Abstract
Monogenic lupus is rare, but its study has contributed immensely to a better understanding of the pathogenesis of systemic lupus erythematosus. The first forms identified were inherited complement deficiencies, which predisposed to lupus due to impaired tolerance, and aberrant clearance of apoptotic bodies and immune complexes. In recent years, several new monogenic disorders with a lupus-like phenotype have been described. These include forms that affect nucleic acid repair, degradation and sensing (TREX1, DNASE1L3), the type I interferon (IFN) pathway (SAMHD1, RNASEH2ABC, ADAR1, IFIH1, ISG15, ACP5, TMEM173) and B cell development checkpoints (PRKCD; RAG2). Pathways informed by these newly described disorders have continued to improve our understanding of systemic lupus erythematosus.
Collapse
Affiliation(s)
- Patricia Costa-Reis
- Pediatrics Department, Hospital de Santa Maria, Lisbon, Portugal; Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
| | - Kathleen E Sullivan
- The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, 3615 Civic Center Blvd., Philadelphia, PA, USA
| |
Collapse
|
57
|
Abstract
PURPOSE OF REVIEW Systemic lupus erythematosus (SLE) is a multisystem autoimmune disease known for its clinical heterogeneity. Over time, new insights into the complex genetic origin of SLE have started to explain some of this clinical variability. These findings, reviewed here, have also yielded important understanding in the immune mechanisms behind SLE pathogenesis. RECENT FINDINGS Several new monogenic disorders with lupus-like phenotype have been described. These can be organized into physiologic pathways that parallel mechanisms of disease in SLE. Examples include genes important for DNA damage repair (e.g., TREX1), nucleic acid sensing and type I interferon overproduction (e.g., STING, TREX1), apoptosis (FASLG), tolerance (PRKCD), and clearance of self-antigen (DNASE1L3). Further study of monogenic lupus may lead to better genotype/phenotype correlations in SLE. Eventually, the ability to understand individual patients according to their genetic profile may allow the development of more targeted and personalized approaches to therapy.
Collapse
Affiliation(s)
- Mindy S Lo
- Division of Immunology, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
58
|
El-Sayed ZA, El-Owaidy RH, Mohamed NL, Shehata BA. Alpha beta double negative T cells in children with systemic lupus erythematosus: The relation to disease activity and characteristics. Mod Rheumatol 2017; 28:654-660. [DOI: 10.1080/14397595.2017.1377146] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Zeinab A. El-Sayed
- Pediatric Allergy and Immunology Unit, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| | - Rasha H. El-Owaidy
- Pediatric Allergy and Immunology Unit, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| | - Neama L. Mohamed
- Clinical Pathology Department, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| | - Beshoy A. Shehata
- Pediatric Allergy and Immunology Unit, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| |
Collapse
|
59
|
Tsokos GC, Lo MS, Costa Reis P, Sullivan KE. New insights into the immunopathogenesis of systemic lupus erythematosus. Nat Rev Rheumatol 2017; 12:716-730. [PMID: 27872476 DOI: 10.1038/nrrheum.2016.186] [Citation(s) in RCA: 783] [Impact Index Per Article: 111.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The aetiology of systemic lupus erythematosus (SLE) is multifactorial, and includes contributions from the environment, stochastic factors, and genetic susceptibility. Great gains have been made in understanding SLE through the use of genetic variant identification, mouse models, gene expression studies, and epigenetic analyses. Collectively, these studies support the concept that defective clearance of immune complexes and biological waste (such as apoptotic cells), neutrophil extracellular traps, nucleic acid sensing, lymphocyte signalling, and interferon production pathways are all central to loss of tolerance and tissue damage. Increased understanding of the pathogenesis of SLE is driving a renewed interest in targeted therapy, and researchers are now on the verge of developing targeted immunotherapy directed at treating either specific organ system involvement or specific subsets of patients with SLE. Accordingly, this Review places these insights within the context of our current understanding of the pathogenesis of SLE and highlights pathways that are ripe for therapeutic targeting.
Collapse
Affiliation(s)
- George C Tsokos
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, 110 Francis Street, Boston, Massachusetts 02215, USA
| | - Mindy S Lo
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
| | - Patricia Costa Reis
- Department of Pediatrics, Lisbon Medical School, Lisbon University, Santa Maria Hospital, Avenida Professor Egas Moniz, 1649-035 Lisbon, Portugal
| | - Kathleen E Sullivan
- Division of Allergy and Immunology, The Children's Hospital of Philadelphia, The University of Pennsylvania Perelman School of Medicine, 3615 Civic Center Boulevard, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
60
|
Yamada A, Arakaki R, Saito M, Kudo Y, Ishimaru N. Dual Role of Fas/FasL-Mediated Signal in Peripheral Immune Tolerance. Front Immunol 2017; 8:403. [PMID: 28424702 PMCID: PMC5380675 DOI: 10.3389/fimmu.2017.00403] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 03/21/2017] [Indexed: 12/20/2022] Open
Abstract
Fas-mediated apoptosis contributes to physiological and pathological cellular processes, such as differentiation and survival. In particular, the roles of Fas in immune cells are complex and critical for the maintenance of immune tolerance. The precise pathways and unique functions associated with Fas/FasL-mediated signaling in the immune system are known. The dual character of Fas/FasL-mediated immune regulation that induces beneficial or harmful effects is associated with the onset or development of immune disorders. Studies on mutations in genes encoding Fas and FasL gene of humans and mice contributed to our understanding of the pathogenesis of autoimmune diseases. Here, we review the opposing functions of Fas/FasL-mediated signaling, bilateral effects of Fas/FasL on in immune cells, and complex pathogenesis of autoimmunity mediated by Fas/FasL.
Collapse
Affiliation(s)
- Akiko Yamada
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Rieko Arakaki
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Masako Saito
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yasusei Kudo
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Naozumi Ishimaru
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| |
Collapse
|
61
|
Weinreich MA, Vogel TP, Rao VK, Milner JD. Up, Down, and All Around: Diagnosis and Treatment of Novel STAT3 Variant. Front Pediatr 2017; 5:49. [PMID: 28349047 PMCID: PMC5347118 DOI: 10.3389/fped.2017.00049] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 02/27/2017] [Indexed: 11/13/2022] Open
Abstract
The number of identified monogenic causes of childhood-onset autoimmunity due to nodal and extranodal lymphoproliferation has increased. These pathogenic genetic variants provide the potential for pathway-specific treatment. Novel variants also require pathway-specific verification. In this report, we describe a 14-year-old patient with a novel variant in STAT3. We report clinical and laboratory findings that support STAT3 p.G419R as a novel pathogenic STAT3 gain-of-function variant.
Collapse
Affiliation(s)
- Michael Alexander Weinreich
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH) , Bethesda, MD , USA
| | - Tiphanie P Vogel
- Center for Human Immunobiology, Texas Children's Hospital, Baylor College of Medicine , Houston, TX , USA
| | - V Koneti Rao
- Division of Intramural Research, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH) , Bethesda, MD , USA
| | - Joshua D Milner
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH) , Bethesda, MD , USA
| |
Collapse
|
62
|
van der Werff ten Bosch J, van den Akker M. Genetic predisposition and hematopoietic malignancies in children: Primary immunodeficiency. Eur J Med Genet 2016; 59:647-653. [DOI: 10.1016/j.ejmg.2016.03.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 02/18/2016] [Accepted: 03/08/2016] [Indexed: 01/24/2023]
|
63
|
Guffroy A, Gies V, Martin M, Korganow AS. [Primary immunodeficiency and autoimmunity]. Rev Med Interne 2016; 38:383-392. [PMID: 27889323 DOI: 10.1016/j.revmed.2016.10.388] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 10/03/2016] [Accepted: 10/20/2016] [Indexed: 12/15/2022]
Abstract
Many evidences highlight that immunodeficiency and autoimmunity are two sides of a same coin. Primary immune deficiencies (PIDs), which are rare mono- or multigenic defects of innate or adaptative immunity, frequently associate with autoimmunity. Analyses of single-gene defects in immune pathways of families with PIDs, by new tools of molecular biology (next genome sequencing technologies), allowed a better understanding of the ways that could both drive immune defect with immune deficiency and autoimmunity. Moreover, genes implicated in rare single-gene defects are now known to be also involved in polygenic conventional autoimmune diseases. Here, we describe the main autoimmune symptoms occurring in PIDs and the underlying mechanisms that lead to autoimmunity in immunodeficiency. We review the links between autoimmunity and immunodeficiency and purpose some principles of care for patients with PIDs and autoimmunity.
Collapse
Affiliation(s)
- A Guffroy
- CNRS UPR 3572 « immunopathologie et chimie thérapeutique », laboratoire d'excellence Medalis, institut de biologie moléculaire et cellulaire (IBMC), 67000 Strasbourg, France; Service d'immunologie clinique et de médecine interne, Centre national de référence des maladies auto-immunes rares, hôpitaux universitaires de Strasbourg, 1, place de l'Hôpital, 67091 Strasbourg, France.
| | - V Gies
- CNRS UPR 3572 « immunopathologie et chimie thérapeutique », laboratoire d'excellence Medalis, institut de biologie moléculaire et cellulaire (IBMC), 67000 Strasbourg, France; Service d'immunologie clinique et de médecine interne, Centre national de référence des maladies auto-immunes rares, hôpitaux universitaires de Strasbourg, 1, place de l'Hôpital, 67091 Strasbourg, France
| | - M Martin
- CNRS UPR 3572 « immunopathologie et chimie thérapeutique », laboratoire d'excellence Medalis, institut de biologie moléculaire et cellulaire (IBMC), 67000 Strasbourg, France; Service d'immunologie clinique et de médecine interne, Centre national de référence des maladies auto-immunes rares, hôpitaux universitaires de Strasbourg, 1, place de l'Hôpital, 67091 Strasbourg, France
| | - A-S Korganow
- CNRS UPR 3572 « immunopathologie et chimie thérapeutique », laboratoire d'excellence Medalis, institut de biologie moléculaire et cellulaire (IBMC), 67000 Strasbourg, France; Service d'immunologie clinique et de médecine interne, Centre national de référence des maladies auto-immunes rares, hôpitaux universitaires de Strasbourg, 1, place de l'Hôpital, 67091 Strasbourg, France
| |
Collapse
|
64
|
Mutations in CRADD Result in Reduced Caspase-2-Mediated Neuronal Apoptosis and Cause Megalencephaly with a Rare Lissencephaly Variant. Am J Hum Genet 2016; 99:1117-1129. [PMID: 27773430 PMCID: PMC5097945 DOI: 10.1016/j.ajhg.2016.09.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 09/19/2016] [Indexed: 12/02/2022] Open
Abstract
Lissencephaly is a malformation of cortical development typically caused by deficient neuronal migration resulting in cortical thickening and reduced gyration. Here we describe a “thin” lissencephaly (TLIS) variant characterized by megalencephaly, frontal predominant pachygyria, intellectual disability, and seizures. Trio-based whole-exome sequencing and targeted re-sequencing identified recessive mutations of CRADD in six individuals with TLIS from four unrelated families of diverse ethnic backgrounds. CRADD (also known as RAIDD) is a death-domain-containing adaptor protein that oligomerizes with PIDD and caspase-2 to initiate apoptosis. TLIS variants cluster in the CRADD death domain, a platform for interaction with other death-domain-containing proteins including PIDD. Although caspase-2 is expressed in the developing mammalian brain, little is known about its role in cortical development. CRADD/caspase-2 signaling is implicated in neurotrophic factor withdrawal- and amyloid-β-induced dendritic spine collapse and neuronal apoptosis, suggesting a role in cortical sculpting and plasticity. TLIS-associated CRADD variants do not disrupt interactions with caspase-2 or PIDD in co-immunoprecipitation assays, but still abolish CRADD’s ability to activate caspase-2, resulting in reduced neuronal apoptosis in vitro. Homozygous Cradd knockout mice display megalencephaly and seizures without obvious defects in cortical lamination, supporting a role for CRADD/caspase-2 signaling in mammalian brain development. Megalencephaly and lissencephaly associated with defective programmed cell death from loss of CRADD function in humans implicate reduced apoptosis as an important pathophysiological mechanism of cortical malformation. Our data suggest that CRADD/caspase-2 signaling is critical for normal gyration of the developing human neocortex and for normal cognitive ability.
Collapse
|
65
|
Zeissig Y, Petersen BS, Franke A, Blumberg RS, Zeissig S. Rare phenotypes in the understanding of autoimmunity. Immunol Cell Biol 2016; 94:943-948. [PMID: 27562064 PMCID: PMC5371426 DOI: 10.1038/icb.2016.76] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 08/16/2016] [Indexed: 12/16/2022]
Abstract
The study of rare phenotypes has a long history in the description of autoimmune disorders. First Mendelian syndromes of idiopathic tissue destruction were defined more than 100 years ago and were later revealed to result from immune-mediated reactivity against self. In the past two decades, continuous advances in sequencing technology and particularly the advent of next-generation sequencing have allowed to define the genetic basis of an ever-growing number of Mendelian forms of autoimmunity. This has provided unique insight into the molecular pathways that govern immunological homeostasis and that are indispensable for the prevention of self-reactive immune-mediated tissue damage and 'horror autotoxicus'. Here we will discuss selected examples of past and recent investigations into rare phenotypes of autoimmunity that have delineated pathways critical for central and peripheral control of the adaptive immune system. We will outline the implications of these findings for rare and common forms of autoimmunity and will discuss the benefits and potential pitfalls of the integration of next-generation sequencing into algorithms for clinical diagnostics. Because of the concise nature of this review, we will focus on syndromes caused by defects in the control of adaptive immunity as innate immune-mediated autoinflammatory disorders have been covered in excellent recent reviews on Mendelian and polygenic forms of autoimmunity.
Collapse
Affiliation(s)
- Yvonne Zeissig
- Department of General Pediatrics, University Medical Center Dresden, Technical University Dresden, Dresden, Germany
| | - Britt-Sabina Petersen
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Richard S Blumberg
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sebastian Zeissig
- Department of Medicine I, University Medical Center Dresden, Technical University Dresden, Dresden, Germany
- Center for Regenerative Therapies, Technical University Dresden, Dresden, Germany
| |
Collapse
|
66
|
Aberdein D, Munday JS, Gandolfi B, Dittmer KE, Malik R, Garrick DJ, Lyons LA. A FAS-ligand variant associated with autoimmune lymphoproliferative syndrome in cats. Mamm Genome 2016; 28:47-55. [DOI: 10.1007/s00335-016-9668-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 10/17/2016] [Indexed: 01/25/2023]
|
67
|
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: 2.0] [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
|
68
|
Rivas-Larrauri F, Yamazaki-Nakashimada MA. Lupus eritematoso sistémico: ¿es una sola enfermedad? ACTA ACUST UNITED AC 2016; 12:274-81. [DOI: 10.1016/j.reuma.2016.01.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 12/21/2015] [Accepted: 01/08/2016] [Indexed: 01/01/2023]
|
69
|
Endoplasmic reticulum-resident E3 ubiquitin ligase Hrd1 controls B-cell immunity through degradation of the death receptor CD95/Fas. Proc Natl Acad Sci U S A 2016; 113:10394-9. [PMID: 27573825 DOI: 10.1073/pnas.1606742113] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Humoral immunity involves multiple checkpoints during B-cell development, maturation, and activation. The cell death receptor CD95/Fas-mediated apoptosis plays a critical role in eliminating the unwanted activation of B cells by self-reactive antigens and in maintaining B-cell homeostasis through activation-induced B-cell death (AICD). The molecular mechanisms controlling AICD remain largely undefined. Herein, we show that the E3 ubiquitin ligase Hrd1 protected B cells from activation-induced cell death by degrading the death receptor Fas. Hrd1-null B cells exhibited high Fas expression during activation and rapidly underwent Fas-mediated apoptosis, which could be largely inhibited by FasL neutralization. Fas mutation in Hrd1 KO mice abrogated the increase in B-cell AICD. We identified Hrd1 as the first E3 ubiquitin ligase of the death receptor Fas and Hrd1-mediated Fas destruction as a molecular mechanism in regulating B-cell immunity.
Collapse
|
70
|
Mistry P, Kaplan MJ. Cell death in the pathogenesis of systemic lupus erythematosus and lupus nephritis. Clin Immunol 2016; 185:59-73. [PMID: 27519955 DOI: 10.1016/j.clim.2016.08.010] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/05/2016] [Accepted: 08/08/2016] [Indexed: 12/12/2022]
Abstract
Nephritis is one of the most severe complications of systemic lupus erythematosus (SLE). One key characteristic of lupus nephritis (LN) is the deposition of immune complexes containing nucleic acids and/or proteins binding to nucleic acids and autoantibodies recognizing these molecules. A variety of cell death processes are implicated in the generation and externalization of modified nuclear autoantigens and in the development of LN. Among these processes, apoptosis, primary and secondary necrosis, NETosis, necroptosis, pyroptosis, and autophagy have been proposed to play roles in tissue damage and immune dysregulation. Cell death occurs in healthy individuals during conditions of homeostasis yet autoimmunity does not develop, at least in part, because of rapid clearance of dying cells. In SLE, accelerated cell death combined with a clearance deficiency may lead to the accumulation and externalization of nuclear autoantigens and to autoantibody production. In addition, specific types of cell death may modify autoantigens and alter their immunogenicity. These modified molecules may then become novel targets of the immune system and promote autoimmune responses in predisposed hosts. In this review, we examine various cell death pathways and discuss how enhanced cell death, impaired clearance, and post-translational modifications of proteins could contribute to the development of lupus nephritis.
Collapse
Affiliation(s)
- Pragnesh Mistry
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
71
|
Perazzio SF, Granados Á, Salomão R, Silva NP, Carneiro-Sampaio M, Andrade LEC. High frequency of immunodeficiency-like states in systemic lupus erythematosus: a cross-sectional study in 300 consecutive patients. Rheumatology (Oxford) 2016; 55:1647-55. [DOI: 10.1093/rheumatology/kew227] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Indexed: 12/29/2022] Open
|
72
|
Otsuki N, Konno T, Kurahashi T, Suzuki S, Lee J, Okada F, Iuchi Y, Homma T, Fujii J. The SOD1 transgene expressed in erythroid cells alleviates fatal phenotype in congenic NZB/NZW-F1 mice. Free Radic Res 2016; 50:793-800. [PMID: 27080108 DOI: 10.1080/10715762.2016.1178388] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Oxidative stress due to a superoxide dismutase 1 (SOD1) deficiency causes anemia and autoimmune responses, which are phenotypically similar to autoimmune hemolytic anemia (AIHA) and systemic lupus erythematosus (SLE) in C57BL/6 mice and aggravates AIHA pathogenesis in New Zealand black (NZB) mice. We report herein on an evaluation of the role of reactive oxygen species (ROS) in a model mouse with inherited SLE, that is, F1 mice of the NZB × New Zealand white (NZW) strain. The ROS levels within red blood cells (RBCs) of the F1 mice were similar to the NZW mice but lower compared to the NZB mice throughout adult period. Regarding SLE pathogenesis, we examined the effects of an SOD1 deficiency or the overexpression of human SOD1 in erythroid cells by establishing corresponding congenic F1 mice. A SOD1 deficiency caused an elevation in ROS production, methemoglobin content, and hyperoxidation of peroxiredoxin in RBC of the F1 mice, which were all consistent with elevated oxidative stress. However, while the overexpression of human SOD1 in erythroid cells extended the life span of the congenic F1 mice, the SOD1 deficiency had no effect on life span compared to wild-type F1 mice. It is generally recognized that NZW mice possess a larval defect in the immune system and that NZB mice trigger an autoimmune reaction in the F1 mice. Our results suggest that the oxidative insult originated from the NZB mouse background has a functional role in triggering an aberrant immune reaction, leading to fatal responses in F1 mice.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Takujiro Homma
- a Department of Biochemistry and Molecular Biology , Graduate School of Medical Science, Yamagata University , Yamagata , Japan
| | | |
Collapse
|
73
|
Huang Y, Deng D, Li H, Xiao Q, Huang L, Zhang B, Ye F, Ye B, Mo Z, Yang X, Liu Z. Fas-670A>G polymorphism is not associated with an increased risk of acute myeloid leukemia development. Biomed Rep 2016; 4:153-160. [PMID: 26893830 PMCID: PMC4734045 DOI: 10.3892/br.2015.564] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 12/07/2015] [Indexed: 11/18/2022] Open
Abstract
The association between the increased risk of acute myeloid leukemia (AML) and Fas promoter polymorphisms has been reported previously; however, the results are inconclusive. The present study performed one case-control study to investigate the association, and a total of 98 AML patients and 2,014 healthy controls were genotyped. The data showed that the distribution of Fas-670AA, GA and GG genotypes among the AML patients were not significantly different from those of the healthy controls, all P>0.05. Following this a sub-study was conducted to analyze individuals who neither smoked nor drank. The results demonstrated that there was still no significant association between the Fas-670 polymorphism and risk of AML development, all P>0.05. Furthermore, in order to address a more accurate estimation of the association, a meta-analysis was conducted. Data were systematically collected from the Pubmed, EMBASE and the Wanfang Library. A total of 3 studies were included in this meta-analysis, which contained 1,144 AML cases and 3,806 controls. No significant association was detected between the Fas-670A>G polymorphism and AML risk [GA+GG vs. AA: odds ratio (OR) 0.93; 95% confidence interval (CI), 0.79–1.09; GG vs. AA: OR, 1.01; 95% CI, 0.82–1.24; GA vs. AA: OR, 1.12; 95% CI, 0.94–1.32; GG vs. AA+GA: OR, 0.94; 95% CI, 0.79–1.12; G vs. A: OR, 1.01; 95% CI, 0.91–1.12; all P>0.05). The analysis clearly indicated that there was no significant connection between the Fas-670A>G polymorphism and the increased risk of AML.
Collapse
Affiliation(s)
- Ying Huang
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Donghong Deng
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Hongying Li
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Qiang Xiao
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Lulu Huang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Bing Zhang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Fanghui Ye
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Bingbing Ye
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Zengnan Mo
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xiaobo Yang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China; Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Zhenfang Liu
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
74
|
Genomics is rapidly advancing precision medicine for immunological disorders. Nat Immunol 2016; 16:1001-4. [PMID: 26382860 DOI: 10.1038/ni.3275] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
75
|
Sobh A, Crestani E, Cangemi B, Kane J, Chou J, Pai SY, Notarangelo LD, Al-Herz W, Geha RS, Massaad MJ. Autoimmune lymphoproliferative syndrome caused by a homozygous FasL mutation that disrupts FasL assembly. J Allergy Clin Immunol 2016; 137:324-327.e2. [PMID: 26456038 DOI: 10.1016/j.jaci.2015.08.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 08/09/2015] [Accepted: 08/12/2015] [Indexed: 01/06/2023]
Affiliation(s)
- Ali Sobh
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Elena Crestani
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Brittney Cangemi
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Jennifer Kane
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Janet Chou
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Sung-Yun Pai
- Division of Hematology-Oncology, Boston Children's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Luigi D Notarangelo
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Waleed Al-Herz
- Allergy and Clinical Immunology Unit, Department of Pediatrics, Al-Sabah Hospital, Kuwait City, Kuwait; Department of Pediatrics, Kuwait University, Kuwait City, Kuwait
| | - Raif S Geha
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Michel J Massaad
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass.
| |
Collapse
|
76
|
Mesenchymal Stem Cell Treatment in Mice Models of Systemic Lupus Erythematosus. STEM CELL BIOLOGY AND REGENERATIVE MEDICINE 2016. [DOI: 10.1007/978-3-319-46733-7_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
77
|
Ruiz-García R, Mora S, Lozano-Sánchez G, Martínez-Lostao L, Paz-Artal E, Ruiz-Contreras J, Anel A, González-Granado LI, Moreno-Pérez D, Allende LM. Decreased activation-induced cell death by EBV-transformed B-cells from a patient with autoimmune lymphoproliferative syndrome caused by a novel FASLG mutation. Pediatr Res 2015; 78:603-8. [PMID: 26334989 DOI: 10.1038/pr.2015.170] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 06/08/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND Autoimmune lymphoproliferative syndrome (ALPS) is a primary immunodeficiency characterized by chronic lymphoproliferation, autoimmune manifestations, expansion of double-negative T-cells, and susceptibility to malignancies. Most cases of ALPS are caused by germline or somatic FAS mutations. We report the case of an ALPS patient due to a novel homozygous Fasligand gene mutation (ALPS-FASLG). METHODS ALPS biomarkers were measured and FASLG mutation was identified. Functional characterization was carried out based on activation-induced cell death (AICD) and cytotoxicity assays. RESULTS This report describes the cases of a patient who presented a severe form of ALPS-FASLG, and his brother who had died due to complications related to ALPS. Moreover, in another family, we present the first case of lymphoma in a patient with ALPS-FASLG. Functional studies showed defective Fasligand-mediated apoptosis, cytotoxicity, and AICD in T-cell blasts. Otherwise, expression of the FASLG gene and corresponding protein was normal, but the shedding of the Fasligand was impaired in T-cells. Additionally, analyzing Epstein-Barr virus (EBV)-transformed B-cells, our results indicate impaired AICD in ALPS-FASLG patients. CONCLUSION Patients with autosomal recessive inheritance of ALPS-FASLG have a severe phenotype and a partial defect in AICD in T- and B-cell lines. The Fasligand could play a key role in immune surveillance preventing malignancy.
Collapse
Affiliation(s)
- Raquel Ruiz-García
- Servicio de Inmunología, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Sergio Mora
- Servicio de Inmunología, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Gema Lozano-Sánchez
- UGC de Pediatría, Hospital Materno-Infantil, Hospital Regional Universitario, Málaga, Spain
| | - Luis Martínez-Lostao
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, Zaragoza, Spain
| | - Estela Paz-Artal
- Servicio de Inmunología, Hospital Universitario 12 de Octubre, Madrid, Spain.,Instituto de Investigación I+12, Madrid, Spain.,Facultad de Medicina, Universidad Complutense y Sección de Inmunología, Universidad San Pablo CEU, Madrid, Spain
| | - Jesús Ruiz-Contreras
- Instituto de Investigación I+12, Madrid, Spain.,Unidad de Inmunodeficiencias, Departamento de Pediatría, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Alberto Anel
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, Zaragoza, Spain
| | - Luis I González-Granado
- Instituto de Investigación I+12, Madrid, Spain.,Unidad de Inmunodeficiencias, Departamento de Pediatría, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - David Moreno-Pérez
- UGC de Pediatría, Hospital Materno-Infantil, Hospital Regional Universitario, Málaga, Spain
| | - Luis M Allende
- Servicio de Inmunología, Hospital Universitario 12 de Octubre, Madrid, Spain.,Instituto de Investigación I+12, Madrid, Spain
| |
Collapse
|
78
|
Nabhani S, Ginzel S, Miskin H, Revel-Vilk S, Harlev D, Fleckenstein B, Hönscheid A, Oommen PT, Kuhlen M, Thiele R, Laws HJ, Borkhardt A, Stepensky P, Fischer U. Deregulation of Fas ligand expression as a novel cause of autoimmune lymphoproliferative syndrome-like disease. Haematologica 2015; 100:1189-98. [PMID: 26113417 DOI: 10.3324/haematol.2014.114967] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 06/19/2015] [Indexed: 12/30/2022] Open
Abstract
Autoimmune lymphoproliferative syndrome is frequently caused by mutations in genes involved in the Fas death receptor pathway, but for 20-30% of patients the genetic defect is unknown. We observed that treatment of healthy T cells with interleukin-12 induces upregulation of Fas ligand and Fas ligand-dependent apoptosis. Consistently, interleukin-12 could not induce apoptosis in Fas ligand-deficient T cells from patients with autoimmune lymphoproliferative syndrome. We hypothesized that defects in the interleukin-12 signaling pathway may cause a similar phenotype as that caused by mutations of the Fas ligand gene. To test this, we analyzed 20 patients with autoimmune lymphoproliferative syndrome of unknown cause by whole-exome sequencing. We identified a homozygous nonsense mutation (c.698G>A, p.R212*) in the interleukin-12/interleukin-23 receptor-component IL12RB1 in one of these patients. The mutation led to IL12RB1 protein truncation and loss of cell surface expression. Interleukin-12 and -23 signaling was completely abrogated as demonstrated by deficient STAT4 phosphorylation and interferon γ production. Interleukin-12-mediated expression of membrane-bound and soluble Fas ligand was lacking and basal expression was much lower than in healthy controls. The patient presented with the classical symptoms of autoimmune lymphoproliferative syndrome: chronic non-malignant, non-infectious lymphadenopathy, splenomegaly, hepatomegaly, elevated numbers of double-negative T cells, autoimmune cytopenias, and increased levels of vitamin B12 and interleukin-10. Sanger sequencing and whole-exome sequencing excluded the presence of germline or somatic mutations in genes known to be associated with the autoimmune lymphoproliferative syndrome. Our data suggest that deficient regulation of Fas ligand expression by regulators such as the interleukin-12 signaling pathway may be an alternative cause of autoimmune lymphoproliferative syndrome-like disease.
Collapse
Affiliation(s)
- Schafiq Nabhani
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Sebastian Ginzel
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany Department of Computer Science, Bonn-Rhein-Sieg University of Applied Sciences, Sankt Augustin, Germany
| | - Hagit Miskin
- Pediatric Hematology Unit, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Shoshana Revel-Vilk
- Department of Pediatric Hematology-Oncology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Dan Harlev
- Pediatric Hematology Unit, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Bernhard Fleckenstein
- Department of Clinical and Molecular Virology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Andrea Hönscheid
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Prasad T Oommen
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Michaela Kuhlen
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Ralf Thiele
- Department of Computer Science, Bonn-Rhein-Sieg University of Applied Sciences, Sankt Augustin, Germany
| | - Hans-Jürgen Laws
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Arndt Borkhardt
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Polina Stepensky
- Department of Pediatric Hematology-Oncology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Ute Fischer
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
79
|
Butt D, Chan TD, Bourne K, Hermes JR, Nguyen A, Statham A, O'Reilly LA, Strasser A, Price S, Schofield P, Christ D, Basten A, Ma CS, Tangye SG, Phan TG, Rao VK, Brink R. FAS Inactivation Releases Unconventional Germinal Center B Cells that Escape Antigen Control and Drive IgE and Autoantibody Production. Immunity 2015; 42:890-902. [PMID: 25979420 DOI: 10.1016/j.immuni.2015.04.010] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 03/09/2015] [Accepted: 03/25/2015] [Indexed: 12/01/2022]
Abstract
The mechanistic links between genetic variation and autoantibody production in autoimmune disease remain obscure. Autoimmune lymphoproliferative syndrome (ALPS) is caused by inactivating mutations in FAS or FASL, with autoantibodies thought to arise through failure of FAS-mediated removal of self-reactive germinal center (GC) B cells. Here we show that FAS is in fact not required for this process. Instead, FAS inactivation led to accumulation of a population of unconventional GC B cells that underwent somatic hypermutation, survived despite losing antigen reactivity, and differentiated into a large population of plasma cells that included autoantibody-secreting clones. IgE(+) plasma cell numbers, in particular, increased after FAS inactivation and a major cohort of ALPS-affected patients were found to have hyper-IgE. We propose that these previously unidentified cells, designated "rogue GC B cells," are a major driver of autoantibody production and provide a mechanistic explanation for the linked production of IgE and autoantibodies in autoimmune disease.
Collapse
Affiliation(s)
- Danyal Butt
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, UNSW Australia, Darlinghurst, NSW 2010, Australia
| | - Tyani D Chan
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, UNSW Australia, Darlinghurst, NSW 2010, Australia
| | - Katherine Bourne
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia
| | - Jana R Hermes
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia
| | - Akira Nguyen
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia
| | - Aaron Statham
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia
| | - Lorraine A O'Reilly
- Molecular Genetics of Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Andreas Strasser
- Molecular Genetics of Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Susan Price
- Molecular Development Section, Laboratory of Immunology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Peter Schofield
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia
| | - Daniel Christ
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, UNSW Australia, Darlinghurst, NSW 2010, Australia
| | - Antony Basten
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, UNSW Australia, Darlinghurst, NSW 2010, Australia
| | - Cindy S Ma
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, UNSW Australia, Darlinghurst, NSW 2010, Australia
| | - Stuart G Tangye
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, UNSW Australia, Darlinghurst, NSW 2010, Australia
| | - Tri Giang Phan
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, UNSW Australia, Darlinghurst, NSW 2010, Australia
| | - V Koneti Rao
- Molecular Development Section, Laboratory of Immunology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Robert Brink
- Immunology Division, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, UNSW Australia, Darlinghurst, NSW 2010, Australia.
| |
Collapse
|
80
|
Crampton SP, Morawski PA, Bolland S. Linking susceptibility genes and pathogenesis mechanisms using mouse models of systemic lupus erythematosus. Dis Model Mech 2015; 7:1033-46. [PMID: 25147296 PMCID: PMC4142724 DOI: 10.1242/dmm.016451] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Systemic lupus erythematosus (SLE) represents a challenging autoimmune disease from a clinical perspective because of its varied forms of presentation. Although broad-spectrum steroids remain the standard treatment for SLE, they have many side effects and only provide temporary relief from the symptoms of the disease. Thus, gaining a deeper understanding of the genetic traits and biological pathways that confer susceptibility to SLE will help in the design of more targeted and effective therapeutics. Both human genome-wide association studies (GWAS) and investigations using a variety of mouse models of SLE have been valuable for the identification of the genes and pathways involved in pathogenesis. In this Review, we link human susceptibility genes for SLE with biological pathways characterized in mouse models of lupus, and discuss how the mechanistic insights gained could advance drug discovery for the disease.
Collapse
Affiliation(s)
- Steve P Crampton
- Laboratory of Immunogenetics, National Institute of Allergic and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Peter A Morawski
- Laboratory of Immunogenetics, National Institute of Allergic and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Silvia Bolland
- Laboratory of Immunogenetics, National Institute of Allergic and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| |
Collapse
|
81
|
Fujii J, Kurahashi T, Konno T, Homma T, Iuchi Y. Oxidative stress as a potential causal factor for autoimmune hemolytic anemia and systemic lupus erythematosus. World J Nephrol 2015; 4:213-222. [PMID: 25949934 PMCID: PMC4419130 DOI: 10.5527/wjn.v4.i2.213] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 01/05/2015] [Accepted: 02/09/2015] [Indexed: 02/05/2023] Open
Abstract
The kidneys and the blood system mutually exert influence in maintaining homeostasis in the body. Because the kidneys control erythropoiesis by producing erythropoietin and by supporting hematopoiesis, anemia is associated with kidney diseases. Anemia is the most prevalent genetic disorder, and it is caused by a deficiency of glucose 6-phosphate dehydrogenase (G6PD), for which sulfhydryl oxidation due to an insufficient supply of NADPH is a likely direct cause. Elevated reactive oxygen species (ROS) result in the sulfhydryl oxidation and hence are another potential cause for anemia. ROS are elevated in red blood cells (RBCs) under superoxide dismutase (SOD1) deficiency in C57BL/6 mice. SOD1 deficient mice exhibit characteristics similar to autoimmune hemolytic anemia (AIHA) and systemic lupus erythematosus (SLE) at the gerontic stage. An examination of AIHA-prone New Zealand Black (NZB) mice, which have normal SOD1 and G6PD genes, indicated that ROS levels in RBCs are originally high and further elevated during aging. Transgenic overexpression of human SOD1 in erythroid cells effectively suppresses ROS elevation and ameliorates AIHA symptoms such as elevated anti-RBC antibodies and premature death in NZB mice. These results support the hypothesis that names oxidative stress as a risk factor for AIHA and other autoimmune diseases such as SLE. Herein we discuss the association between oxidative stress and SLE pathogenesis based mainly on the genetic and phenotypic characteristics of NZB and New Zealand white mice and provide insight into the mechanism of SLE pathogenesis.
Collapse
|
82
|
Lundy SK, Klinker MW. Characterization and activity of Fas ligand producing CD5⁺ B cells. Methods Mol Biol 2015; 1190:81-102. [PMID: 25015275 DOI: 10.1007/978-1-4939-1161-5_7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
B lymphocytes make several contributions to immune regulation including production of antibodies with regulatory properties, release of immune suppressive cytokines, and expression of death-inducing ligands. A role for Fas ligand (FasL)-expressing "killer" B cells in regulating T helper cell survival and chronic inflammation has been demonstrated in animal models of schistosome worm infection, asthma, and autoimmune arthritis. Interestingly, a population of CD5(+) B cells found in the spleen and lungs of naïve mice constitutively expresses FasL and has potent killer function against T helper cells that is antigen-specific and FasL-dependent. Killer B cells therefore represent a novel target for immune modulation in many disease settings. Our laboratory has recently published methods of characterizing FasL(+) B cells and inducing their proliferation in vitro. This chapter will describe detailed methods of identifying and expanding killer B cells from mice, detecting FasL expression in B cells, and performing functional killing assays against antigen-specific TH cells.
Collapse
Affiliation(s)
- Steven K Lundy
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, 4043 Biomedical Sciences Research Bldg., 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA,
| | | |
Collapse
|
83
|
A mutation in caspase-9 decreases the expression of BAFFR and ICOS in patients with immunodeficiency and lymphoproliferation. Genes Immun 2015; 16:151-61. [PMID: 25569260 DOI: 10.1038/gene.2014.74] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 10/09/2014] [Accepted: 11/17/2014] [Indexed: 01/25/2023]
Abstract
Lymphocyte apoptosis is mainly induced by either death receptor-dependent activation of caspase-8 or mitochondria-dependent activation of caspase-9. Mutations in caspase-8 lead to autoimmunity/lymphoproliferation and immunodeficiency. This work describes a heterozygous H237P mutation in caspase-9 that can lead to similar disorders. H237P mutation was detected in two patients: Pt1 with autoimmunity/lymphoproliferation, severe hypogammaglobulinemia and Pt2 with mild hypogammaglobulinemia and Burkitt lymphoma. Their lymphocytes displayed defective caspase-9 activity and decreased apoptotic and activation responses. Transfection experiments showed that mutant caspase-9 display defective enzyme and proapoptotic activities and a dominant-negative effect on wild-type caspase-9. Ex vivo analysis of the patients' lymphocytes and in vitro transfection experiments showed that the expression of mutant caspase-9 correlated with a downregulation of BAFFR (B-cell-activating factor belonging to the TNF family (BAFF) receptor) in B cells and ICOS (inducible T-cell costimulator) in T cells. Both patients carried a second inherited heterozygous mutation missing in the relatives carrying H237P: Pt1 in the transmembrane activator and CAML interactor (TACI) gene (S144X) and Pt2 in the perforin (PRF1) gene (N252S). Both mutations have been previously associated with immunodeficiencies in homozygosis or compound heterozygosis. Taken together, these data suggest that caspase-9 mutations may predispose to immunodeficiency by cooperating with other genetic factors, possibly by downregulating the expression of BAFFR and ICOS.
Collapse
|
84
|
Casanova JL, Conley ME, Seligman SJ, Abel L, Notarangelo LD. Guidelines for genetic studies in single patients: lessons from primary immunodeficiencies. ACTA ACUST UNITED AC 2014; 211:2137-49. [PMID: 25311508 PMCID: PMC4203950 DOI: 10.1084/jem.20140520] [Citation(s) in RCA: 180] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Casanova and colleagues discuss the importance of single-patient genetic studies in the discovery of novel primary immunodeficiencies and offer insight into the standards and criteria that should accompany these studies. Can genetic and clinical findings made in a single patient be considered sufficient to establish a causal relationship between genotype and phenotype? We report that up to 49 of the 232 monogenic etiologies (21%) of human primary immunodeficiencies (PIDs) were initially reported in single patients. The ability to incriminate single-gene inborn errors in immunodeficient patients results from the relative ease in validating the disease-causing role of the genotype by in-depth mechanistic studies demonstrating the structural and functional consequences of the mutations using blood samples. The candidate genotype can be causally connected to a clinical phenotype using cellular (leukocytes) or molecular (plasma) substrates. The recent advent of next generation sequencing (NGS), with whole exome and whole genome sequencing, induced pluripotent stem cell (iPSC) technology, and gene editing technologies—including in particular the clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 technology—offer new and exciting possibilities for the genetic exploration of single patients not only in hematology and immunology but also in other fields. We propose three criteria for deciding if the clinical and experimental data suffice to establish a causal relationship based on only one case. The patient’s candidate genotype must not occur in individuals without the clinical phenotype. Experimental studies must indicate that the genetic variant impairs, destroys, or alters the expression or function of the gene product (or two genetic variants for compound heterozygosity). The causal relationship between the candidate genotype and the clinical phenotype must be confirmed via a relevant cellular phenotype, or by default via a relevant animal phenotype. When supported by satisfaction of rigorous criteria, the report of single patient–based discovery of Mendelian disorders should be encouraged, as it can provide the first step in the understanding of a group of human diseases, thereby revealing crucial pathways underlying physiological and pathological processes.
Collapse
Affiliation(s)
- Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065 Howard Hughes Medical Institute, New York, NY 10065 Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France Paris Descartes University, Imagine Institute, 75015 Paris, France Pediatric Hematology-Immunology Unit, Necker Hospital for Sick Children, 75015 Paris, France
| | - Mary Ellen Conley
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065
| | - Stephen J Seligman
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY 10595
| | - Laurent Abel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065 Howard Hughes Medical Institute, New York, NY 10065 Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, 75015 Paris, France Paris Descartes University, Imagine Institute, 75015 Paris, France
| | - Luigi D Notarangelo
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115 Department of Pediatrics and Pathology, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
85
|
Loss of the death receptor CD95 (Fas) expression by dendritic cells protects from a chronic viral infection. Proc Natl Acad Sci U S A 2014; 111:8559-64. [PMID: 24912151 DOI: 10.1073/pnas.1401750111] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Chronic viral infections incapacitate adaptive immune responses by "exhausting" virus-specific T cells, inducing their deletion and reducing productive T-cell memory. Viral infection rapidly induces death receptor CD95 (Fas) expression by dendritic cells (DCs), making them susceptible to elimination by the immune response. Lymphocytic choriomeningitis virus (LCMV) clone 13, which normally establishes a chronic infection, is rapidly cleared in C57Black6/J mice with conditional deletion of Fas in DCs. The immune response to LCMV is characterized by an extended survival of virus-specific effector T cells. Moreover, transfer of Fas-negative DCs from noninfected mice to preinfected animals results in either complete clearance of the virus or a significant reduction of viral titers. Thus, DC-specific Fas expression plays a role in regulation of antiviral responses and suggests a strategy for stimulation of T cells in chronically infected animals and humans to achieve the clearance of persistent viruses.
Collapse
|
86
|
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: 15.0] [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
|
87
|
Abstract
Genetics unquestionably contributes to systemic lupus erythematosus (SLE) predisposition, progression and outcome. Nevertheless, single-gene defects causing lupus-like phenotypes have been infrequently documented. The majority of the identified genetic SLE risk factors are, therefore, common variants, responsible for a small effect on the global risk. Recently, genome wide association studies led to the identification of a growing number of gene variants associated with SLE susceptibility, particular disease phenotypes, and antibody profiles. Further studies addressed the biological effects of these variants. In addition, the role of epigenetics has recently been revealed. These combined efforts contributed to a better understanding of SLE pathogenesis and to the characterization of clinically relevant pathways. In this review, we describe SLE-associated single-gene defects, common variants, and epigenetic changes. We also discuss the limitations of current methods and the challenges that we still have to face in order to incorporate genomic and epigenomic data into clinical practice.
Collapse
|
88
|
Abstract
Paediatric-onset systemic lupus erythematosus (SLE) is usually more severe than its adult counterpart. In particular, there is a higher incidence of renal and central nervous system involvement. Specific measures to assess disease activity and damage have been implemented. The disease is very rare before the fifth birthday and therefore the onset of an SLE picture in the first years of life should lead to the suspicion of the presence of one of the rare monogenic diseases that causes SLE or of one of those congenital diseases that has been showed to be closely associated with the SLE.
Collapse
Affiliation(s)
- Clara Malattia
- Department of Pediatrics, University of Genoa and Pediatria e Reumatologia, G. Gaslini Institute, Genoa, Italy.
| | | |
Collapse
|
89
|
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
|
90
|
Association of FAS and FAS ligand genes polymorphism and risk of systemic lupus erythematosus. ScientificWorldJournal 2013; 2013:176741. [PMID: 24348139 PMCID: PMC3848338 DOI: 10.1155/2013/176741] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Accepted: 09/17/2013] [Indexed: 02/07/2023] Open
Abstract
FAS/FASL pathway plays a critical role in maintaining peripheral immune tolerance; therefore, the apoptosis genes, Fas and Fas ligand (FasL), could be suitable candidate genes in human SLE susceptibility. Materials and Methods. In this case-control study, 106 SLE patients and 149 sex, age, and ethnicity matched healthy controls were genotyped for the Fas A-670G and FasLC-844T polymorphisms by polymerase chain reaction-restriction fragment length polymorphism method (PCR-RFLP). Results. The frequency of -670AA genotype was significantly higher in SLE patients than control group and the risk of SLE was 2.1-fold greater in subjects with AA genotype (P = 0.03). The frequency of -670A allele was significantly higher in SLE patients than in controls too (58% versus 49%, P = 0.03). The -844CC genotype frequency was significantly higher in SLE patients than in healthy controls and the risk of SLE was 2.8-fold greater in these subjects (P = 0.01). The C allele frequency was significantly higher in patients than in controls (69% versus 49%, P = 0.001). Increased SLE risk was observed in individuals with combined effect of Fas-670AA and FasL-844CC genotypes (P = 0.001). Conclusion. Fas-670AA and FasL-844CC genotypes were associated with SLE risk, and combined effect of -670AA and -844CC genotypes might increase SLE susceptibility.
Collapse
|
91
|
Selective depletion of FOXP3(high) cells by Fas-Fas-L-induced apoptosis occurs in CD4(+)CD25(+)-enriched populations during repeated expansion. Cytotherapy 2013; 15:1286-96. [PMID: 23993302 DOI: 10.1016/j.jcyt.2013.05.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 05/20/2013] [Accepted: 05/28/2013] [Indexed: 11/23/2022]
Abstract
BACKGROUND AIMS Expansion of anti-CD25 bead-isolated human Tregs culture has paradoxically resulted in reduced suppressive activity, but the mechanism(s) responsible for these observations are poorly defined. METHODS Magnetic-bead isolated human CD25(+) cells were expanded with anti-CD3/CD28 beads and high doses of rhIL-2. Detection of Fas and Fas ligand (Fas-L) expression, activation of Caspase 8, cell proliferation and cytokine production was evaluated by multi-color fluorescence-activated cell sorting analysis. The role of Fas-Fas-L-mediated cell death was dissected through the use of agonist or antagonist monoclonal antibodies directed at Fas and Fas-L. RESULTS Repeated expansion of bead-enriched CD4(+)CD25(+) cells generated a cellular product with markedly reduced suppressive activity and with significantly increased CD8(+) T cells and CD4(+) T cells producing interferon-γ and/or interleukin-2. We showed that Fas-Fas-L-mediated apoptosis of CD4(+)FOXP3(high) cells and rapid cell-cycling of CD8(+) T cells were collectively responsible for the reduced proportion of CD4(+)FOXP3(high) cells in expanded cultures. The depletion of CD4(+)FOXP3(high) cells and activation of Caspase 8 in CD4(+)FOXP3(high) cells was attenuated by Fas antagonist antibody, ZB4, in short-term culture. However, the loss of CD4(+)FOXP3(high) cells during expansion was not prevented by either Fas or Fas-L antagonist antibodies. CONCLUSIONS Taken together, the data show that Fas-Fas-L-mediated apoptosis may limit the expansion of anti-CD25 bead-isolated cells in vitro.
Collapse
|
92
|
Belot A, Kasher PR, Trotter EW, Foray AP, Debaud AL, Rice GI, Szynkiewicz M, Zabot MT, Rouvet I, Bhaskar SS, Daly SB, Dickerson JE, Mayer J, O’Sullivan J, Juillard L, Urquhart JE, Fawdar S, Marusiak AA, Stephenson N, Waszkowycz B, Beresford MW, Biesecker LG, Black GCM, René C, Eliaou JF, Fabien N, Ranchin B, Cochat P, Gaffney PM, Rozenberg F, Lebon P, Malcus C, Crow YJ, Brognard J, Bonnefoy N. Protein kinase cδ deficiency causes mendelian systemic lupus erythematosus with B cell-defective apoptosis and hyperproliferation. ARTHRITIS AND RHEUMATISM 2013; 65:2161-71. [PMID: 23666743 PMCID: PMC4066615 DOI: 10.1002/art.38008] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 05/02/2013] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is a prototype autoimmune disease that is assumed to occur via a complex interplay of environmental and genetic factors. Rare causes of monogenic SLE have been described, providing unique insights into fundamental mechanisms of immune tolerance. The aim of this study was to identify the cause of an autosomal-recessive form of SLE. METHODS We studied 3 siblings with juvenile-onset SLE from 1 consanguineous kindred and used next-generation sequencing to identify mutations in the disease-associated gene. We performed extensive biochemical, immunologic, and functional assays to assess the impact of the identified mutations on B cell biology. RESULTS We identified a homozygous missense mutation in PRKCD, encoding protein kinase δ (PKCδ), in all 3 affected siblings. Mutation of PRKCD resulted in reduced expression and activity of the encoded protein PKCδ (involved in the deletion of autoreactive B cells), leading to resistance to B cell receptor- and calcium-dependent apoptosis and increased B cell proliferation. Thus, as for mice deficient in PKCδ, which exhibit an SLE phenotype and B cell expansion, we observed an increased number of immature B cells in the affected family members and a developmental shift toward naive B cells with an immature phenotype. CONCLUSION Our findings indicate that PKCδ is crucial in regulating B cell tolerance and preventing self-reactivity in humans, and that PKCδ deficiency represents a novel genetic defect of apoptosis leading to SLE.
Collapse
Affiliation(s)
- Alexandre Belot
- Centre de Référence des Maladies Rénales Rares, Hospices Civils de Lyon, INSERM U1111, UMS3444/US8, Université Claude Bernard Lyon 1, and Université de Lyon, Lyon, France
| | - Paul R. Kasher
- Manchester Academic Health Science Centre and University of Manchester, Manchester, UK
| | - Eleanor W. Trotter
- Paterson Institute for Cancer Research and University of Manchester, Manchester, UK
| | - Anne-Perrine Foray
- Hospices Civils de Lyon, INSERM U1111, UMS3444/US8, Université Claude Bernard Lyon 1, and Université de Lyon, Lyon, France
| | - Anne-Laure Debaud
- INSERM U1111, UMS3444/US8, Université Claude Bernard Lyon 1, and Université de Lyon, Lyon, France
| | - Gillian I. Rice
- Manchester Academic Health Science Centre and University of Manchester, Manchester, UK
| | - Marcin Szynkiewicz
- Manchester Academic Health Science Centre and University of Manchester, Manchester, UK
| | - Marie-Therese Zabot
- Centre de Biotechnologie Cellulaire, Groupement Hospitalier Est, and Hospices Civils de Lyon, Lyon, France
| | - Isabelle Rouvet
- Centre de Biotechnologie Cellulaire, Groupement Hospitalier Est, and Hospices Civils de Lyon, Lyon, France
| | - Sanjeev S. Bhaskar
- Manchester Academic Health Science Centre and University of Manchester, Manchester, UK
| | - Sarah B. Daly
- Manchester Academic Health Science Centre and University of Manchester, Manchester, UK
| | - Jonathan E. Dickerson
- Manchester Academic Health Science Centre and University of Manchester, Manchester, UK
| | - Josephine Mayer
- Manchester Academic Health Science Centre and University of Manchester, Manchester, UK
| | - James O’Sullivan
- Manchester Academic Health Science Centre and University of Manchester, Manchester, UK
| | - Laurent Juillard
- Hôpital E. Herriot, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, and Université de Lyon, Lyon, France
| | - Jill E. Urquhart
- Manchester Academic Health Science Centre and University of Manchester, Manchester, UK
| | - Shameem Fawdar
- Paterson Institute for Cancer Research and University of Manchester, Manchester, UK
| | - Anna A. Marusiak
- Paterson Institute for Cancer Research and University of Manchester, Manchester, UK
| | - Natalie Stephenson
- Paterson Institute for Cancer Research and University of Manchester, Manchester, UK
| | - Bohdan Waszkowycz
- Paterson Institute for Cancer Research and University of Manchester, Manchester, UK
| | | | - Leslie G. Biesecker
- NIH, Bethesda, Maryland, and NIH Intramural Sequencing Center, Rockville, Maryland
| | - Graeme C. M. Black
- Manchester Academic Health Science Centre and University of Manchester, Manchester, UK
| | - Céline René
- Centre Hospitalier Régional Universitaire de Montpellier and Université Montpellier 1, Montpellier, France
| | - Jean-François Eliaou
- Centre Hospitalier Régional Universitaire de Montpellier, Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U896, Université Montpellier 1, and Institut Régional du Cancer de Montpellier, Montpellier, Montpellier, France
| | - Nicole Fabien
- Centre Hospitalier Lyon Sud and Hospices Civils de Lyon, Lyon, France
| | - Bruno Ranchin
- Centre de Référence des Maladies Rénales Rares and Hospices Civils de Lyon, Lyon, France
| | - Pierre Cochat
- Centre de Référence des Maladies Rénales Rares, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Université de Lyon, and Epidemiologie Pharmacologie Investigation Clinique Information Medicale Mere Enfant (EPICIME), Lyon, France
| | | | | | | | | | - Yanick J. Crow
- Manchester Academic Health Science Centre and University of Manchester, Manchester, UK
| | - John Brognard
- Paterson Institute for Cancer Research and University of Manchester, Manchester, UK
| | - Nathalie Bonnefoy
- Hospices Civils de Lyon, INSERM U1111, UMS3444/US8, Université Claude Bernard Lyon 1, and Université de Lyon, Lyon, France, and Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U896, Université Montpellier 1, and Institut Régional du Cancer de Montpellier, Montpellier, France
| |
Collapse
|
93
|
Aricò M, Boggio E, Cetica V, Melensi M, Orilieri E, Clemente N, Cappellano G, Buttini S, Soluri MF, Comi C, Dufour C, Pende D, Dianzani I, Ellis SR, Pagliano S, Marcenaro S, Ramenghi U, Chiocchetti A, Dianzani U. Variations of the UNC13D gene in patients with autoimmune lymphoproliferative syndrome. PLoS One 2013; 8:e68045. [PMID: 23840885 PMCID: PMC3698121 DOI: 10.1371/journal.pone.0068045] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Accepted: 05/24/2013] [Indexed: 01/08/2023] Open
Abstract
Autoimmune lymphoproliferative syndrome (ALPS) is caused by genetic defects decreasing Fas function and is characterized by lymphadenopathy/splenomegaly and expansion of CD4/CD8 double-negative T cells. This latter expansion is absent in the ALPS variant named Dianzani Autoimmune/lymphoproliferative Disease (DALD). In addition to the causative mutations, the genetic background influences ALPS and DALD development. We previously suggested a disease-modifying role for the perforin gene involved in familial hemophagocytic lymphohistiocytosis (FHL). The UNC13D gene codes for Munc13-4, which is involved in perforin secretion and FHL development, and thus, another candidate for a disease-modifying role in ALPS and DALD. In this work, we sequenced UNC13D in 21 ALPS and 20 DALD patients and compared these results with sequences obtained from 61 healthy subjects and 38 multiple sclerosis (MS) patients. We detected four rare missense variations in three heterozygous ALPS patients carrying p.Cys112Ser, p.Val781Ile, and a haplotype comprising both p.Ile848Leu and p.Ala995Pro. Transfection of the mutant cDNAs into HMC-1 cells showed that they decreased granule exocytosis, compared to the wild-type construct. An additional rare missense variation, p.Pro271Ser, was detected in a healthy subject, but this variation did not decrease Munc13-4 function. These data suggest that rare loss-of-function variations of UND13D are risk factors for ALPS development.
Collapse
Affiliation(s)
- Maurizio Aricò
- Department of Pediatric Hematology Oncology, Meyer Children Hospital, Firenze, Italy
| | - Elena Boggio
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), “A. Avogadro” University of Eastern Piedmont, Novara, Italy
- Department of Health Sciences, “A. Avogadro” University of Eastern Piedmont, Novara, Italy
| | - Valentina Cetica
- Department of Pediatric Hematology Oncology, Meyer Children Hospital, Firenze, Italy
| | - Matteo Melensi
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), “A. Avogadro” University of Eastern Piedmont, Novara, Italy
- Department of Health Sciences, “A. Avogadro” University of Eastern Piedmont, Novara, Italy
| | - Elisabetta Orilieri
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), “A. Avogadro” University of Eastern Piedmont, Novara, Italy
- Department of Health Sciences, “A. Avogadro” University of Eastern Piedmont, Novara, Italy
| | - Nausicaa Clemente
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), “A. Avogadro” University of Eastern Piedmont, Novara, Italy
- Department of Health Sciences, “A. Avogadro” University of Eastern Piedmont, Novara, Italy
| | - Giuseppe Cappellano
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), “A. Avogadro” University of Eastern Piedmont, Novara, Italy
- Department of Health Sciences, “A. Avogadro” University of Eastern Piedmont, Novara, Italy
| | - Sara Buttini
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), “A. Avogadro” University of Eastern Piedmont, Novara, Italy
- Department of Translational Medicine, "A. Avogadro" University of Eastern Piedmont, Novara, Italy
| | - Maria Felicia Soluri
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), “A. Avogadro” University of Eastern Piedmont, Novara, Italy
- Department of Health Sciences, “A. Avogadro” University of Eastern Piedmont, Novara, Italy
| | - Cristoforo Comi
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), “A. Avogadro” University of Eastern Piedmont, Novara, Italy
- Department of Translational Medicine, "A. Avogadro" University of Eastern Piedmont, Novara, Italy
| | | | | | - Irma Dianzani
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), “A. Avogadro” University of Eastern Piedmont, Novara, Italy
- Department of Health Sciences, “A. Avogadro” University of Eastern Piedmont, Novara, Italy
| | - Steven R. Ellis
- Department of Biochemistry and Molecular Biology, University of Louisville, Louisville, Kentucky, United States of America
| | - Sara Pagliano
- Department of Pediatrics, University of Torino, Torino, Italy
| | | | - Ugo Ramenghi
- Department of Pediatrics, University of Torino, Torino, Italy
| | - Annalisa Chiocchetti
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), “A. Avogadro” University of Eastern Piedmont, Novara, Italy
- Department of Health Sciences, “A. Avogadro” University of Eastern Piedmont, Novara, Italy
- * E-mail:
| | - Umberto Dianzani
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), “A. Avogadro” University of Eastern Piedmont, Novara, Italy
- Department of Health Sciences, “A. Avogadro” University of Eastern Piedmont, Novara, Italy
| |
Collapse
|
94
|
Abstract
PURPOSE OF REVIEW The recognition that apoptosis - programmed cell death - is an important mechanism in immune homeostasis has led to the identification of human disorders associated with defects in the critical control mechanism. RECENT FINDINGS Patients have been identified with defects affecting the extrinsic apoptotic pathway mediated by the protein receptor FAS which results in the autoimmune lymphoproliferative syndrome and more recently in defects affecting the intrinsic apoptotic pathway mediated by RAS proteins resulting in the RAS-associated autoimmune leukoproliferative disorder. This review summarizes the immunopathogenesis, clinical features and diagnostic approaches to these human disorders. SUMMARY Apoptotic pathways are critical in the maintenance of leukocyte homeostasis, and genetic defects impacting these can result in clinical disease manifested as expansion of selected leukocyte populations, autoimmunity, increased risk for malignancy and in some situations defects in host defense.
Collapse
|
95
|
Somatic loss of heterozygosity, but not haploinsufficiency alone, leads to full-blown autoimmune lymphoproliferative syndrome in 1 of 12 family members with FAS start codon mutation. Clin Immunol 2013; 147:61-68. [PMID: 23524443 DOI: 10.1016/j.clim.2013.02.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 01/02/2013] [Accepted: 02/15/2013] [Indexed: 12/19/2022]
Abstract
We describe a family with 12 members carrying a heterozygous germline FAS c.3G>T start codon mutation leading to FAS haploinsufficiency. One patient had autoimmune lymphoproliferative syndrome (ALPS), one had recovered from ALPS, and ten mutation-positive relatives (MPRs) were healthy. FAS-mediated apoptosis and surface expression of FAS in single-positive T cells were lower for MPRs but did not discriminate between them and the ALPS patient. However, double-negative (DN) T cells of the ALPS patient had no FAS expression due to somatic loss of heterozygosity. Our results in this kindred suggest that FAS haploinsufficiency does not cause ALPS-FAS, but that modifying genetic events are crucial for its pathogenesis. FAS surface expression on DN T cells should be assessed routinely and FAS haploinsufficient patients should be followed as its potential for lymphomagenesis is not well defined and a second hit might occur later on.
Collapse
|
96
|
Sahin SB, Cetinkalp S, Erdogan M, Yilmaz C, Berdeli A. Fas, Fas Ligand, and vitamin D Receptor FokI gene polymorphisms in patients with type 1 diabetes mellitus in the Aegean region of Turkey. Genet Test Mol Biomarkers 2012; 16:1179-83. [PMID: 22946667 DOI: 10.1089/gtmb.2012.0173] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVE Several gene polymorphisms have been reported to be associated with the risk of developing type 1 diabetes. Among them, the human leukocyte antigen locus is the strongest genetic determinant. To identify additional genetic markers, we aimed to evaluate the relationship between the Fas, Fas ligand (FasL), and vitamin D receptor (VDR) FokI gene polymorphisms and the susceptibility to type 1 diabetes in the Aegean region of Turkey. MATERIALS AND METHODS Eighty-five patients with type 1 diabetes and 80 healthy controls were included in this study. The Fas -670A/G, FasL -843C/T, and VDR FokI gene polymorphisms were evaluated using the polymerase chain reaction-restriction fragment length polymorphism method. RESULTS The evaluation of the Fas genotype and the gene allele frequency did not show statistically significant differences between the patient and control group. Distribution of the FasL genotype differed significantly between patients and controls. The distribution of the VDR FokI genotype and allele frequencies differed significantly between the patients and controls. Individuals with type 1 diabetes presented less commonly with the FokI f allele. CONCLUSIONS Our findings suggest that the FasL -843C/T and VDR FokI gene polymorphisms are associated with type 1 diabetes in the Agean region of Turkey; however, the Fas -670A/G gene polymorphism is not.
Collapse
Affiliation(s)
- Serap Baydur Sahin
- Department of Endocrinology and Metabolism Disease, Ege University Medical School, Bornova, Izmir, Turkey.
| | | | | | | | | |
Collapse
|
97
|
Belot A, Cimaz R. Monogenic forms of systemic lupus erythematosus: new insights into SLE pathogenesis. Pediatr Rheumatol Online J 2012; 10:21. [PMID: 22883345 PMCID: PMC3489560 DOI: 10.1186/1546-0096-10-21] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Accepted: 07/29/2012] [Indexed: 01/23/2023] Open
Abstract
The pathogenesis of Systemic Lupus Erythematosus (SLE) is complex and remains poorly understood. Infectious triggers, genetic background, immunological abnormalities and environmental factors are all supposed to interact for the disease development. Familial SLE as well as early-onset juvenile SLE studies make it possible to identify monogenic causes of SLE. Identification of these rare inherited conditions is of great interest to understand both SLE pathogenesis and molecular human tolerance mechanisms. Complement deficiencies, genetic overproduction of interferon-α and apoptosis defects are the main situations that can lead to monogenic SLE.Here, we review the different genes involved in monogenic SLE and highlight their importance in SLE pathogenesis.
Collapse
Affiliation(s)
- Alexandre Belot
- Pediatric nephrology and rheumatology Unit, Hôpital Femme Mère Enfant, Lyon, Bron, Université de Lyon, Lyon, CNRS UMR5239, France.
| | - Rolando Cimaz
- Rheumatology Unit, Anna Meyer Children's Hospital, University of Florence, Florence, Italy
| |
Collapse
|
98
|
Magerus-Chatinet A, Stolzenberg MC, Lanzarotti N, Neven B, Daussy C, Picard C, Neveux N, Desai M, Rao M, Ghosh K, Madkaikar M, Fischer A, Rieux-Laucat F. Autoimmune lymphoproliferative syndrome caused by a homozygous null FAS ligand (FASLG) mutation. J Allergy Clin Immunol 2012; 131:486-90. [PMID: 22857792 DOI: 10.1016/j.jaci.2012.06.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 06/07/2012] [Accepted: 06/12/2012] [Indexed: 11/17/2022]
Abstract
BACKGROUND Autoimmune lymphoproliferative syndrome (ALPS) is characterized by chronic nonmalignant lymphoproliferation, accumulation of double-negative T cells, hypergammaglobulinemia G and A, and autoimmune cytopenia. OBJECTIVES Although mostly associated with FAS mutations, different genetic defects leading to impaired apoptosis have been described in patients with ALPS, including the FAS ligand gene (FASLG) in rare cases. Here we report on the first case of complete FAS ligand deficiency caused by a homozygous null mutant. METHODS Double-negative T-cell counts and plasma IL-10 and FAS ligand concentrations were determined as ALPS markers. The FASLG gene was sequenced, and its expression was analyzed by means of Western blotting. FAS ligand function was assessed based on reactivation-induced cell death. RESULTS We describe a patient born to consanguineous parents who presented with a severe form of ALPS caused by FASLG deficiency. Although the clinical presentation was compatible with a homozygous FAS mutation, FAS-induced apoptosis was normal, and plasma FAS ligand levels were not detectable. This patient carries a homozygous, germline, single-base-pair deletion in FASLG exon 1, leading to a premature stop codon (F87fs x95) and a complete defect in FASLG expression. The healthy parents were each heterozygous for the mutation, confirming its recessive trait. CONCLUSION FAS ligand deficiency should be screened in patients presenting with ALPS features but lacking the usual markers, including plasma soluble FAS ligand and an in vitro apoptotic defect. An activation-induced cell death test could help in discrimination.
Collapse
|
99
|
Increased protection from vaccinia virus infection in mice genetically prone to lymphoproliferative disorders. J Virol 2012; 86:6010-22. [PMID: 22438562 DOI: 10.1128/jvi.07176-11] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Mutations in the genes that encode Fas or Fas ligand (FasL) can result in poor restraints on lymphocyte activation and in increased susceptibility to autoimmune disorders. Because these mutations portend a continuously activated immune state, we hypothesized that they might in some cases confer resistance to infection. To examine this possibility, the immune response to, morbidity caused by, and clearance of vaccinia virus (VACV) Western Reserve was examined in 5- to 7-week-old Fas mutant (lpr) mice, before an overt lymphoproliferative disorder was observable. On day 6 after VACV infection, C57BL/6-lpr (B6-lpr) mice had decreased morbidity, decreased viral titers, and an increased percentage and number of CD4(+) and CD8(+) T cells. As early as day 2 after infection, B6-lpr mice had decreased liver and spleen viral titers and increased numbers of and increased gamma interferon (IFN-γ) production by several different effector cell populations. Depletion of individual effector cell subsets did not inhibit the resistance of B6-lpr mice. Uninfected B6-lpr mice also had increased numbers of NK cells, γδ(+) T cells, and CD44(+) CD4(+) and CD44(+) CD8(+) T cells compared to uninfected B6 mice. Antibody to IFN-γ resulted in increased virus load in both B6 and B6-lpr mice and eliminated the differences in viral titers between them. These results suggest that IFN-γ produced by multiple activated leukocyte populations in Fas-deficient hosts enhances resistance to some viral infections.
Collapse
|
100
|
The -346T polymorphism of the SH2D1A gene is a risk factor for development of autoimmunity/lymphoproliferation in males with defective Fas function. Hum Immunol 2012; 73:585-92. [PMID: 22425739 DOI: 10.1016/j.humimm.2012.02.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Revised: 02/03/2012] [Accepted: 02/27/2012] [Indexed: 11/20/2022]
Abstract
Inherited defects decreasing function of the Fas death receptor cause autoimmune lymphoproliferative syndrome (ALPS) and its variant Dianzani autoimmune lymphoproliferative disease (DALD). Since a deleterious mutation of the SH2D1A gene protects MRLlpr/lpr mice from ALPS development, we investigated the role of SH2D1A, located in the X chromosome, in 51 patients with ALPS or DALD by mutational screening of coding and regulative sequences. Allelic frequency of the -346C>T polymorphism was different in male patients and controls (-346T: 61% vs 36%, p = 0.01), with similar frequencies in ALPS and DALD. By contrast, no differences were found among females or between the controls and patients with multiple sclerosis (229 males, 157 females). Further analyses showed that -346C was a methylation site in CD8(+) T and natural killer cells, and SH2D1A expression was higher in -346T than in -346C males. Finally, in vitro-activated T cells from -346T males produced lower amounts of interferon-γ than those from -346C males. These data suggest that -346T is a predisposing factor for ALPS and DALD in males possibly because of its effect on SAP expression influencing the T-cell response.
Collapse
|