Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 171(2): 385-397.e11, 2017 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-28919076

RESUMEN

T cell receptor (TCR) signaling without CD28 can elicit primary effector T cells, but memory T cells generated during this process are anergic, failing to respond to secondary antigen exposure. We show that, upon T cell activation, CD28 transiently promotes expression of carnitine palmitoyltransferase 1a (Cpt1a), an enzyme that facilitates mitochondrial fatty acid oxidation (FAO), before the first cell division, coinciding with mitochondrial elongation and enhanced spare respiratory capacity (SRC). microRNA-33 (miR33), a target of thioredoxin-interacting protein (TXNIP), attenuates Cpt1a expression in the absence of CD28, resulting in cells that thereafter are metabolically compromised during reactivation or periods of increased bioenergetic demand. Early CD28-dependent mitochondrial engagement is needed for T cells to remodel cristae, develop SRC, and rapidly produce cytokines upon restimulation-cardinal features of protective memory T cells. Our data show that initial CD28 signals during T cell activation prime mitochondria with latent metabolic capacity that is essential for future T cell responses.


Asunto(s)
Antígenos CD28/metabolismo , Activación de Linfocitos , Mitocondrias/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología , Animales , Carnitina O-Palmitoiltransferasa , Inhibidores Enzimáticos/farmacología , Compuestos Epoxi/farmacología , Humanos , Interleucina-15/inmunología , Ratones , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T/metabolismo , Estrés Fisiológico , Linfocitos T/metabolismo
2.
J Allergy Clin Immunol ; 153(1): 67-76, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37977527

RESUMEN

Chronic nonmalignant lymphoproliferation and autoimmune cytopenia are relevant manifestations of immunohematologic diseases of childhood. Their diagnostic classification is challenging but important for therapy. Autoimmune lymphoproliferative syndrome (ALPS) is a genetically defined inborn error of immunity combining these manifestations, but it can explain only a small proportion of cases. Diagnostic categories such as ALPS-like disease, common variable immunodeficiency, or Evans syndrome have therefore been used. Advances in genetics and increasing availablity of targeted therapies call for more therapy-oriented disease classification. Moreover, recent discoveries in the (re)analysis of genetic conditions affecting FAS signaling ask for a more precise definition of ALPS. In this review, we propose the term autoimmune lymphoproliferative immunodeficiencies for a disease phenotype that is enriched for patients with genetic diseases for which targeted therapies are available. For patients without a current molecular diagnosis, this term defines a subgroup of immune dysregulatory disorders for further studies. Within the concept of autoimmune lymphoproliferative immunodeficiencies, we propose a revision of the ALPS classification, restricting use of this term to conditions with clear evidence of perturbation of FAS signaling and resulting specific biologic and clinical consequences. This proposed approach to redefining ALPS and other lymphoproliferative conditions provides a framework for disease classification and diagnosis that is relevant for the many specialists confronted with these diseases.


Asunto(s)
Anemia Hemolítica Autoinmune , Enfermedades Autoinmunes , Síndrome Linfoproliferativo Autoinmune , Inmunodeficiencia Variable Común , Enfermedades del Sistema Inmune , Trastornos Linfoproliferativos , Humanos , Síndrome Linfoproliferativo Autoinmune/diagnóstico , Síndrome Linfoproliferativo Autoinmune/genética , Síndrome Linfoproliferativo Autoinmune/terapia , Fenotipo , Receptor fas/genética , Trastornos Linfoproliferativos/diagnóstico , Trastornos Linfoproliferativos/genética , Trastornos Linfoproliferativos/terapia
3.
J Allergy Clin Immunol ; 153(1): 203-215, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37793571

RESUMEN

BACKGROUND: The autoimmune lymphoproliferative syndrome (ALPS) is a noninfectious and nonmalignant lymphoproliferative disease frequently associated with autoimmune cytopenia resulting from defective FAS signaling. We previously described germline monoallelic FAS (TNFRSF6) haploinsufficient mutations associated with somatic events, such as loss of heterozygosity on the second allele of FAS, as a cause of ALPS-FAS. These somatic events were identified by sequencing FAS in DNA from double-negative (DN) T cells, the pathognomonic T-cell subset in ALPS, in which the somatic events accumulated. OBJECTIVE: We sought to identify whether a somatic event affecting the FAS-associated death domain (FADD) gene could be related to the disease onset in 4 unrelated patients with ALPS carrying a germline monoallelic mutation of the FADD protein inherited from a healthy parent. METHODS: We sequenced FADD and performed array-based comparative genomic hybridization using DNA from sorted CD4+ or DN T cells. RESULTS: We found homozygous FADD mutations in the DN T cells from all 4 patients, which resulted from uniparental disomy. FADD deficiency caused by germline heterozygous FADD mutations associated with a somatic loss of heterozygosity was a phenocopy of ALPS-FAS without the more complex symptoms reported in patients with germline biallelic FADD mutations. CONCLUSIONS: The association of germline and somatic events affecting the FADD gene is a new genetic cause of ALPS.


Asunto(s)
Síndrome Linfoproliferativo Autoinmune , Proteína de Dominio de Muerte Asociada a Fas , Humanos , Apoptosis/genética , Enfermedades Autoinmunes/genética , Síndrome Linfoproliferativo Autoinmune/genética , Hibridación Genómica Comparativa , ADN , Receptor fas/genética , Proteína de Dominio de Muerte Asociada a Fas/genética , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Células Germinativas/patología , Mutación
4.
J Allergy Clin Immunol ; 153(1): 297-308.e12, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37979702

RESUMEN

BACKGROUND: Elevated TCRαß+CD4-CD8- double-negative T cells (DNT) and serum biomarkers help identify FAS mutant patients with autoimmune lymphoproliferative syndrome (ALPS). However, in some patients with clinical features and biomarkers consistent with ALPS, germline or somatic FAS mutations cannot be identified on standard exon sequencing (ALPS-undetermined: ALPS-U). OBJECTIVE: We sought to explore whether complex genetic alterations in the FAS gene escaping standard sequencing or mutations in other FAS pathway-related genes could explain these cases. METHODS: Genetic analysis included whole FAS gene sequencing, copy number variation analysis, and sequencing of FAS cDNA and other FAS pathway-related genes. It was guided by FAS expression analysis on CD57+DNT, which can predict somatic loss of heterozygosity (sLOH). RESULTS: Nine of 16 patients with ALPS-U lacked FAS expression on CD57+DNT predicting heterozygous "loss-of-expression" FAS mutations plus acquired somatic second hits in the FAS gene, enriched in DNT. Indeed, 7 of 9 analyzed patients carried deep intronic mutations or large deletions in the FAS gene combined with sLOH detectable in DNT; 1 patient showed a FAS exon duplication. Three patients had reduced FAS expression, and 2 of them harbored mutations in the FAS promoter, which reduced FAS expression in reporter assays. Three of the 4 ALPS-U patients with normal FAS expression carried heterozygous FADD mutations with sLOH. CONCLUSION: A combination of serum biomarkers and DNT phenotyping is an accurate means to identify patients with ALPS who are missed by routine exome sequencing.


Asunto(s)
Síndrome Linfoproliferativo Autoinmune , Receptor fas , Humanos , Síndrome Linfoproliferativo Autoinmune/diagnóstico , Síndrome Linfoproliferativo Autoinmune/genética , Biomarcadores , Variaciones en el Número de Copia de ADN , Secuenciación del Exoma , Receptor fas/genética , Proteína de Dominio de Muerte Asociada a Fas/genética , Mutación
5.
J Allergy Clin Immunol ; 151(5): 1391-1401.e7, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36621650

RESUMEN

BACKGROUND: Fas ligand (FasL) is expressed by activated T cells and induces death in target cells upon binding to Fas. Loss-of-function FAS or FASLG mutations cause autoimmune-lymphoproliferative syndrome (ALPS) characterized by expanded double-negative T cells (DNT) and elevated serum biomarkers. While most ALPS patients carry heterozygous FAS mutations, FASLG mutations are rare and usually biallelic. Only 2 heterozygous variants were reported, associated with an atypical clinical phenotype. OBJECTIVE: We revisited the significance of heterozygous FASLG mutations as a cause of ALPS. METHODS: Clinical features and biomarkers were analyzed in 24 individuals with homozygous or heterozygous FASLG variants predicted to be deleterious. Cytotoxicity assays were performed with patient T cells and biochemical assays with recombinant FasL. RESULTS: Homozygous FASLG variants abrogated cytotoxicity and resulted in early-onset severe ALPS with elevated DNT, raised vitamin B12, and usually no soluble FasL. In contrast, heterozygous variants affected FasL function by reducing expression, impairing trimerization, or preventing Fas binding. However, they were not associated with elevated DNT and vitamin B12, and they did not affect FasL-mediated cytotoxicity. The dominant-negative effects of previously published variants could not be confirmed. Even Y166C, causing loss of Fas binding with a dominant-negative effect in biochemical assays, did not impair cellular cytotoxicity or cause vitamin B12 and DNT elevation. CONCLUSION: Heterozygous loss-of-function mutations are better tolerated for FASLG than for FAS, which may explain the low frequency of ALPS-FASLG.


Asunto(s)
Síndrome Linfoproliferativo Autoinmune , Humanos , Síndrome Linfoproliferativo Autoinmune/genética , Proteína Ligando Fas/genética , Mutación , Biomarcadores , Vitaminas , Receptor fas/genética , Apoptosis/genética
6.
J Allergy Clin Immunol ; 152(2): 500-516, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37004747

RESUMEN

BACKGROUND: Biallelic mutations in LIG4 encoding DNA-ligase 4 cause a rare immunodeficiency syndrome manifesting as infant-onset life-threatening and/or opportunistic infections, skeletal malformations, radiosensitivity and neoplasia. LIG4 is pivotal during DNA repair and during V(D)J recombination as it performs the final DNA-break sealing step. OBJECTIVES: This study explored whether monoallelic LIG4 missense mutations may underlie immunodeficiency and autoimmunity with autosomal dominant inheritance. METHODS: Extensive flow-cytometric immune-phenotyping was performed. Rare variants of immune system genes were analyzed by whole exome sequencing. DNA repair functionality and T-cell-intrinsic DNA damage tolerance was tested with an ensemble of in vitro and in silico tools. Antigen-receptor diversity and autoimmune features were characterized by high-throughput sequencing and autoantibody arrays. Reconstitution of wild-type versus mutant LIG4 were performed in LIG4 knockout Jurkat T cells, and DNA damage tolerance was subsequently assessed. RESULTS: A novel heterozygous LIG4 loss-of-function mutation (p.R580Q), associated with a dominantly inherited familial immune-dysregulation consisting of autoimmune cytopenias, and in the index patient with lymphoproliferation, agammaglobulinemia, and adaptive immune cell infiltration into nonlymphoid organs. Immunophenotyping revealed reduced naive CD4+ T cells and low TCR-Vα7.2+ T cells, while T-/B-cell receptor repertoires showed only mild alterations. Cohort screening identified 2 other nonrelated patients with the monoallelic LIG4 mutation p.A842D recapitulating clinical and immune-phenotypic dysregulations observed in the index family and displaying T-cell-intrinsic DNA damage intolerance. Reconstitution experiments and molecular dynamics simulations categorize both missense mutations as loss-of-function and haploinsufficient. CONCLUSIONS: This study provides evidence that certain monoallelic LIG4 mutations may cause human immune dysregulation via haploinsufficiency.


Asunto(s)
ADN Ligasas , Síndromes de Inmunodeficiencia , Humanos , ADN Ligasas/genética , Autoinmunidad/genética , Haploinsuficiencia , ADN Ligasa (ATP)/genética , Síndromes de Inmunodeficiencia/genética , Mutación , ADN
7.
J Allergy Clin Immunol ; 151(4): 1081-1095, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36228738

RESUMEN

BACKGROUND: In 2014, germline signal transducer and activator of transcription (STAT) 3 gain-of-function (GOF) mutations were first described to cause a novel multisystem disease of early-onset lymphoproliferation and autoimmunity. OBJECTIVE: This pivotal cohort study defines the scope, natural history, treatment, and overall survival of a large global cohort of patients with pathogenic STAT3 GOF variants. METHODS: We identified 191 patients from 33 countries with 72 unique mutations. Inclusion criteria included symptoms of immune dysregulation and a biochemically confirmed germline heterozygous GOF variant in STAT3. RESULTS: Overall survival was 88%, median age at onset of symptoms was 2.3 years, and median age at diagnosis was 12 years. Immune dysregulatory features were present in all patients: lymphoproliferation was the most common manifestation (73%); increased frequencies of double-negative (CD4-CD8-) T cells were found in 83% of patients tested. Autoimmune cytopenias were the second most common clinical manifestation (67%), followed by growth delay, enteropathy, skin disease, pulmonary disease, endocrinopathy, arthritis, autoimmune hepatitis, neurologic disease, vasculopathy, renal disease, and malignancy. Infections were reported in 72% of the cohort. A cellular and humoral immunodeficiency was observed in 37% and 51% of patients, respectively. Clinical symptoms dramatically improved in patients treated with JAK inhibitors, while a variety of other immunomodulatory treatment modalities were less efficacious. Thus far, 23 patients have undergone bone marrow transplantation, with a 62% survival rate. CONCLUSION: STAT3 GOF patients present with a wide array of immune-mediated disease including lymphoproliferation, autoimmune cytopenias, and multisystem autoimmunity. Patient care tends to be siloed, without a clear treatment strategy. Thus, early identification and prompt treatment implementation are lifesaving for STAT3 GOF syndrome.


Asunto(s)
Enfermedades del Sistema Inmune , Síndromes de Inmunodeficiencia , Niño , Humanos , Autoinmunidad/genética , Estudios de Cohortes , Mutación con Ganancia de Función , Síndromes de Inmunodeficiencia/genética , Mutación , Factor de Transcripción STAT3/genética , Proliferación Celular , Linfocitos
8.
Blood ; 136(9): 1055-1066, 2020 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-32518946

RESUMEN

Molecular dissection of inborn errors of immunity can help to elucidate the nonredundant functions of individual genes. We studied 3 children with an immune dysregulation syndrome of susceptibility to infection, lymphadenopathy, hepatosplenomegaly, developmental delay, autoimmunity, and lymphoma of B-cell (n = 2) or T-cell (n = 1) origin. All 3 showed early autologous T-cell reconstitution following allogeneic hematopoietic stem cell transplantation. By whole-exome sequencing, we identified rare homozygous germline missense or nonsense variants in a known epigenetic regulator of gene expression: ten-eleven translocation methylcytosine dioxygenase 2 (TET2). Mutated TET2 protein was absent or enzymatically defective for 5-hydroxymethylating activity, resulting in whole-blood DNA hypermethylation. Circulating T cells showed an abnormal immunophenotype including expanded double-negative, but depleted follicular helper, T-cell compartments and impaired Fas-dependent apoptosis in 2 of 3 patients. Moreover, TET2-deficient B cells showed defective class-switch recombination. The hematopoietic potential of patient-derived induced pluripotent stem cells was skewed toward the myeloid lineage. These are the first reported cases of autosomal-recessive germline TET2 deficiency in humans, causing clinically significant immunodeficiency and an autoimmune lymphoproliferative syndrome with marked predisposition to lymphoma. This disease phenotype demonstrates the broad role of TET2 within the human immune system.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , Mutación de Línea Germinal , Mutación con Pérdida de Función , Trastornos Linfoproliferativos/genética , Proteínas Proto-Oncogénicas/deficiencia , Inmunodeficiencia Combinada Grave/genética , Aloinjertos , Apoptosis , Subgrupos de Linfocitos B/patología , Técnicas de Reprogramación Celular , Codón sin Sentido , Metilación de ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Dioxigenasas , Resultado Fatal , Femenino , Trasplante de Células Madre Hematopoyéticas , Humanos , Células Madre Pluripotentes Inducidas/patología , Recién Nacido , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/patología , Linfoma de Células T Periférico/genética , Linfoma de Células T Periférico/patología , Masculino , Mutación Missense , Neoplasias Primarias Múltiples/genética , Linaje , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/fisiología , Inmunodeficiencia Combinada Grave/patología , Subgrupos de Linfocitos T/patología , Secuenciación del Exoma
9.
Clin Immunol ; 210: 108316, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31770611

RESUMEN

Germline STAT3 gain-of-function (GOF) mutations have been linked to poly-autoimmunity and lymphoproliferation with variable expressivity and incomplete penetrance. Here we studied the impact of 17 different STAT3 GOF mutations on the canonical STAT3 signaling pathway and correlated the molecular results with clinical manifestations. The mutations clustered in three groups. Group 1 mutants showed altered STAT3 phosphorylation kinetics and strong basal transcriptional activity. They were associated with the highest penetrance of lymphoproliferation and autoimmunity. Group 2 mutants showed a strongly inducible transcriptional reporter activity and were clinically less penetrant. Group 3 mutants were mostly located in the DNA binding domain and showed the strongest DNA binding affinity despite a poor transcriptional reporter response. Thus, the GOF effect of STAT3 mutations is determined by a heterogeneous response pattern at the molecular level. The correlation of response pattern and clinical penetrance indicates a significant contribution of mutation-determined effects on disease manifestations.


Asunto(s)
Mutación con Ganancia de Función/genética , Linfocitos/inmunología , Factor de Transcripción STAT3/genética , Autoinmunidad/genética , Proliferación Celular , Femenino , Humanos , Masculino , Familia de Multigenes , Penetrancia , Fosforilación , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/genética
10.
Eur J Immunol ; 47(2): 364-373, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27925643

RESUMEN

Hemophagocytic lymphohistiocytosis (HLH) is a life-threatening inflammatory syndrome characterized by hyperactivation of lymphocytes and histiocytes. T cells play a key role in HLH pathogenesis, but their differentiation pattern is not well characterized in patients with active HLH. We compared T-cell activation patterns between patients with familial HLH (1°HLH), 2°HLH without apparent infectious trigger (2°HLH) and 2°HLH induced by a viral infection (2°V-HLH). Polyclonal CD8+ T cells are highly activated in 1°HLH and 2°V-HLH, but less in 2°HLH as assessed by HLA-DR expression and marker combination with CD45RA, CCR7, CD127, PD-1 and CD57. Absence of increased HLA-DR expression on T cells excluded active 1° HLH with high sensitivity and specificity. A high proportion of polyclonal CD127- CD4+ T cells expressing HLA-DR, CD57, and perforin is a signature of infants with 1°HLH, much less prominent in virus-associated 2°HLH. The similar pattern and extent of CD8+ T-cell activation compared to 2° V-HLH is compatible with a viral trigger of 1°HLH. However, in most 1°HLH patients no triggering infection was documented and the unique activation of cytotoxic CD4+ T cells indicates that the overall T-cell response in 1°HLH is different. This may reflect different pathways of pathogenesis of these two HLH variants.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfohistiocitosis Hemofagocítica/inmunología , Virosis/inmunología , Antígenos CD57/metabolismo , Diferenciación Celular , Células Cultivadas , Femenino , Antígenos HLA-DR/metabolismo , Humanos , Lactante , Recién Nacido , Activación de Linfocitos , Linfohistiocitosis Hemofagocítica/diagnóstico , Linfohistiocitosis Hemofagocítica/etiología , Masculino , Perforina/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Virosis/complicaciones , Virosis/diagnóstico
11.
Immunol Cell Biol ; 96(10): 1060-1071, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29790605

RESUMEN

Premature T-cell immunosenescence with CD57+ CD8+ T-cell accumulation has been linked to immunodeficiency and autoimmunity in primary immunodeficiencies including activated PI3 kinase delta syndrome (APDS). To address whether CD57 marks the typical senescent T-cell population seen in adult individuals or identifies a distinct population in APDS, we compared CD57+ CD8+ T cells from mostly pediatric APDS patients to those of healthy adults with similarly prominent senescent T cells. CD57+ CD8+ T cells from APDS patients were less differentiated with more CD27+ CD28+ effector memory T cells showing increased PD1 and Eomesodermin expression. In addition, transition of naïve to CD57+ CD8+ T cells was not associated with the characteristic telomere shortening. Nevertheless, they showed the increased interferon-gamma secretion, enhanced degranulation and reduced in vitro proliferation typical of senescent CD57+ CD8+ T cells. Thus, hyperactive PI3 kinase signaling favors premature accumulation of a CD57+ CD8+ T-cell population, which shows most functional features of typical senescent T cells, but is different in terms of differentiation and relative telomere shortening. Initial observations indicate that this specific differentiation state may offer the opportunity to revert premature T-cell immunosenescence and its potential contribution to inflammation and immunodeficiency in APDS.


Asunto(s)
Antígenos CD57/metabolismo , Diferenciación Celular , Síndromes de Inmunodeficiencia/etiología , Síndromes de Inmunodeficiencia/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Acortamiento del Telómero , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Senescencia Celular/genética , Senescencia Celular/inmunología , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Citocinas/metabolismo , Humanos , Inmunofenotipificación , Recuento de Linfocitos , Enfermedades de Inmunodeficiencia Primaria , Sirolimus/farmacología , Subgrupos de Linfocitos T/efectos de los fármacos
12.
Blood ; 127(18): 2193-202, 2016 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-26907631

RESUMEN

Fas is a transmembrane receptor involved in the maintenance of tolerance and immune homeostasis. In murine models, it has been shown to be essential for deletion of autoreactive B cells in the germinal center. The role of Fas in human B-cell selection and in development of autoimmunity in patients carrying FAS mutations is unclear. We analyzed patients with either a somatic FAS mutation or a germline FAS mutation and somatic loss-of-heterozygosity, which allows comparing the fate of B cells with impaired vs normal Fas signaling within the same individual. Class-switched memory B cells showed: accumulation of FAS-mutated B cells; failure to enrich single V, D, J genes and single V-D, D-J gene combinations of the B-cell receptor variable region; increased frequency of variable regions with higher content of positively charged amino acids; and longer CDR3 and maintenance of polyreactive specificities. Importantly, Fas-deficient switched memory B cells showed increased rates of somatic hypermutation. Our data uncover a defect in B-cell selection in patients with FAS mutations, which has implications for the understanding of the pathogenesis of autoimmunity and lymphomagenesis of autoimmune lymphoproliferative syndrome.


Asunto(s)
Síndrome Linfoproliferativo Autoinmune/inmunología , Subgrupos de Linfocitos B/inmunología , Selección Clonal Mediada por Antígenos , Mutación , Receptor fas/fisiología , Apoptosis , Autoinmunidad , Línea Celular Transformada , Transformación Celular Neoplásica , Niño , Codón sin Sentido , Femenino , Mutación del Sistema de Lectura , Mutación de Línea Germinal , Heterocigoto , Humanos , Memoria Inmunológica , Pérdida de Heterocigocidad , Masculino , Análisis de Secuencia de ADN , Hipermutación Somática de Inmunoglobulina , Recombinación V(D)J , Receptor fas/deficiencia , Receptor fas/genética
13.
Blood ; 128(2): 227-38, 2016 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-27099149

RESUMEN

Autoimmune lymphoproliferative syndrome (ALPS) is a human disorder characterized by defective Fas signaling, resulting in chronic benign lymphoproliferation and accumulation of TCRαß(+) CD4(-) CD8(-) double-negative T (DNT) cells. Although their phenotype resembles that of terminally differentiated or exhausted T cells, lack of KLRG1, high eomesodermin, and marginal T-bet expression point instead to a long-lived memory state with potent proliferative capacity. Here we show that despite their terminally differentiated phenotype, human ALPS DNT cells exhibit substantial mitotic activity in vivo. Notably, hyperproliferation of ALPS DNT cells is associated with increased basal and activation-induced phosphorylation of serine-threonine kinases Akt and mechanistic target of rapamycin (mTOR). The mTOR inhibitor rapamycin abrogated survival and proliferation of ALPS DNT cells, but not of CD4(+) or CD8(+) T cells in vitro. In vivo, mTOR inhibition reduced proliferation and abnormal differentiation by DNT cells. Importantly, increased mitotic activity and hyperactive mTOR signaling was also observed in recently defined CD4(+) or CD8(+) precursor DNT cells, and mTOR inhibition specifically reduced these cells in vivo, indicating abnormal programming of Fas-deficient T cells before the DNT stage. Thus, our results identify the mTOR pathway as a major regulator of lymphoproliferation and aberrant differentiation in ALPS.


Asunto(s)
Síndrome Linfoproliferativo Autoinmune/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Transducción de Señal/inmunología , Serina-Treonina Quinasas TOR/inmunología , Adolescente , Adulto , Síndrome Linfoproliferativo Autoinmune/patología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Niño , Preescolar , Femenino , Humanos , Lectinas Tipo C/inmunología , Antígenos Comunes de Leucocito/inmunología , Masculino , Proteínas Proto-Oncogénicas c-akt/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Inmunológicos , Transactivadores/inmunología
14.
J Allergy Clin Immunol ; 139(2): 607-620.e15, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27477328

RESUMEN

BACKGROUND: Patients with heterozygous germline mutations in phosphatase and tensin homolog deleted on chromosome 10 (PTEN) experience autoimmunity and lymphoid hyperplasia. OBJECTIVES: Because regulation of the phosphoinositide 3-kinase (PI3K) pathway is critical for maintaining regulatory T (Treg) cell functions, we investigate Treg cells in patients with heterozygous germline PTEN mutations (PTEN hamartoma tumor syndrome [PHTS]). METHODS: Patients with PHTS were assessed for immunologic conditions, lymphocyte subsets, forkhead box P3 (FOXP3)+ Treg cell levels, and phenotype. To determine the functional importance of phosphatases that control the PI3K pathway, we assessed Treg cell induction in vitro, mitochondrial depolarization, and recruitment of PTEN to the immunologic synapse. RESULTS: Autoimmunity and peripheral lymphoid hyperplasia were found in 43% of 79 patients with PHTS. Immune dysregulation in patients with PHTS included lymphopenia, CD4+ T-cell reduction, and changes in T- and B-cell subsets. Although total CD4+FOXP3+ Treg cell numbers are reduced, frequencies are maintained in the blood and intestine. Despite pathogenic PTEN mutations, the FOXP3+ T cells are phenotypically normal. We show that the phosphatase PH domain leucine-rich repeat protein phosphatase (PHLPP) downstream of PTEN is highly expressed in normal human Treg cells and provides complementary phosphatase activity. PHLPP is indispensable for the differentiation of induced Treg cells in vitro and Treg cell mitochondrial fitness. PTEN and PHLPP form a phosphatase network that is polarized at the immunologic synapse. CONCLUSION: Heterozygous loss of function of PTEN in human subjects has a significant effect on T- and B-cell immunity. Assembly of the PTEN-PHLPP phosphatase network allows coordinated phosphatase activities at the site of T-cell receptor activation, which is important for limiting PI3K hyperactivation in Treg cells despite PTEN haploinsufficiency.


Asunto(s)
Linfocitos B/fisiología , Síndrome de Hamartoma Múltiple/inmunología , Sinapsis Inmunológicas/metabolismo , Subgrupos Linfocitarios/fisiología , Proteínas Nucleares/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Linfocitos T Reguladores/fisiología , Adolescente , Adulto , Anciano , Autoinmunidad , Células Cultivadas , Niño , Factores de Transcripción Forkhead/metabolismo , Síndrome de Hamartoma Múltiple/genética , Humanos , Hiperplasia , Masculino , Potencial de la Membrana Mitocondrial , Persona de Mediana Edad , Mutación/genética , Fosfohidrolasa PTEN/genética , Unión Proteica , Transporte de Proteínas , Transducción de Señal , Adulto Joven
15.
Clin Immunol ; 180: 97-99, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28478106

RESUMEN

A patient with autoimmune lymphoproliferative disorder (ALPS) developed IgG4-related disease. In retrospect, he had high levels of serum IgG4 for several years prior to presenting with IgG4-related pancreatitis. These high IgG4 levels were masked by hypergammaglobulinemia, a common feature of ALPS. We next screened 18 ALPS patients; four of them displayed increased levels of IgG4. Hence, IgG4-related disease should be considered in ALPS patients, especially in those manifesting lymphocytic organ infiltration or excessive hypergammaglobulinaemia. Screening of IgG4-related disease patients for ALPS-associated mutations would provide further information on whether this disease could be a late-onset atypical presentation of ALPS.


Asunto(s)
Síndrome Linfoproliferativo Autoinmune/inmunología , Inmunoglobulina G/inmunología , Adulto , Síndrome Linfoproliferativo Autoinmune/sangre , Síndrome Linfoproliferativo Autoinmune/patología , Humanos , Hipergammaglobulinemia/inmunología , Inmunoglobulina G/sangre , Ganglios Linfáticos/patología , Trastornos Linfoproliferativos/inmunología , Masculino , Páncreas/patología
16.
Blood ; 125(5): 753-61, 2015 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-25414442

RESUMEN

Autoimmune cytopenia is a frequent manifestation of primary immunodeficiencies. Two siblings presented with Evans syndrome, viral infections, and progressive leukopenia. DNA available from one patient showed a homozygous frameshift mutation in tripeptidyl peptidase II (TPP2) abolishing protein expression. TPP2 is a serine exopeptidase involved in extralysosomal peptide degradation. Its deficiency in mice activates cell death programs and premature senescence. Similar to cells from naïve, uninfected TPP2-deficient mice, patient cells showed increased major histocompatibility complex I expression and most CD8(+) T-cells had a senescent CCR7-CD127(-)CD28(-)CD57(+) phenotype with poor proliferative responses and enhanced staurosporine-induced apoptosis. T-cells showed increased expression of the effector molecules perforin and interferon-γ with high expression of the transcription factor T-bet. Age-associated B-cells with a CD21(-) CD11c(+) phenotype expressing T-bet were increased in humans and mice, combined with antinuclear antibodies. Moreover, markers of senescence were also present in human and murine TPP2-deficient fibroblasts. Telomere lengths were normal in patient fibroblasts and granulocytes, and low normal in lymphocytes, which were compatible with activation of stress-induced rather than replicative senescence programs. TPP2 deficiency is the first primary immunodeficiency linking premature immunosenescence to severe autoimmunity. Determination of senescent lymphocytes should be part of the diagnostic evaluation of children with refractory multilineage cytopenias.


Asunto(s)
Envejecimiento/inmunología , Aminopeptidasas/inmunología , Anemia Hemolítica Autoinmune/genética , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/inmunología , Mutación del Sistema de Lectura , Síndromes de Inmunodeficiencia/genética , Serina Endopeptidasas/inmunología , Trombocitopenia/genética , Aminopeptidasas/deficiencia , Aminopeptidasas/genética , Anemia Hemolítica Autoinmune/complicaciones , Anemia Hemolítica Autoinmune/inmunología , Anemia Hemolítica Autoinmune/patología , Animales , Apoptosis , Secuencia de Bases , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Niño , Preescolar , Consanguinidad , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/deficiencia , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/genética , Femenino , Fibroblastos/inmunología , Fibroblastos/metabolismo , Fibroblastos/patología , Expresión Génica , Humanos , Síndromes de Inmunodeficiencia/complicaciones , Síndromes de Inmunodeficiencia/inmunología , Síndromes de Inmunodeficiencia/patología , Masculino , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Perforina/genética , Perforina/inmunología , Serina Endopeptidasas/deficiencia , Serina Endopeptidasas/genética , Hermanos , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología , Trombocitopenia/complicaciones , Trombocitopenia/inmunología , Trombocitopenia/patología
17.
Blood ; 126(14): 1658-69, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26289640

RESUMEN

Omenn syndrome (OS) is a severe immunodeficiency associated with erythroderma, lymphoproliferation, elevated IgE, and hyperactive oligoclonal T cells. A restricted T-cell repertoire caused by defective thymic T-cell development and selection, lymphopenia with homeostatic proliferation, and lack of regulatory T cells are considered key factors in OS pathogenesis. We report 2 siblings presenting with cytomegalovirus (CMV) and Pneumocystis jirovecii infections and recurrent sepsis; one developed all clinical features of OS. Both carried homozygous germline mutations in CARD11 (p.Cys150*), impairing NF-κB signaling and IL-2 production. A somatic second-site mutation reverting the stop codon to a missense mutation (p.Cys150Leu) was detected in tissue-infiltrating T cells of the OS patient. Expression of p.Cys150Leu in CARD11-deficient T cells largely reconstituted NF-κB signaling. The reversion likely occurred in a prethymic T-cell precursor, leading to a chimeric T-cell repertoire. We speculate that in our patient the functional advantage of the revertant T cells in the context of persistent CMV infection, combined with lack of regulatory T cells, may have been sufficient to favor OS. This first observation of OS in a patient with a T-cell activation defect suggests that severely defective T-cell development or homeostatic proliferation in a lymphopenic environment are not required for this severe immunopathology.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/genética , Guanilato Ciclasa/genética , Activación de Linfocitos/genética , Mutación , Inmunodeficiencia Combinada Grave/genética , Linfocitos T Reguladores/inmunología , Animales , Proteínas Adaptadoras de Señalización CARD/deficiencia , Proteínas Adaptadoras de Señalización CARD/inmunología , Femenino , Citometría de Flujo , Guanilato Ciclasa/deficiencia , Guanilato Ciclasa/inmunología , Humanos , Immunoblotting , Inmunohistoquímica , Inmunofenotipificación , Lactante , Activación de Linfocitos/inmunología , Masculino , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Inmunodeficiencia Combinada Grave/inmunología , Hermanos
19.
N Engl J Med ; 369(26): 2504-14, 2013 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-24369075

RESUMEN

BACKGROUND: Severe combined immunodeficiency (SCID) comprises a heterogeneous group of heritable deficiencies of humoral and cell-mediated immunity. Many patients with SCID have lymphocyte-activation defects that remain uncharacterized. METHODS: We performed genetic studies in four patients, from four families of Northern Cree ancestry, who had clinical characteristics of SCID, including early onset of severe viral, bacterial, and fungal infections despite normal B-cell and T-cell counts. Genomewide homozygosity mapping was used to identify a candidate region, which was found on chromosome 8; all genes within this interval were sequenced. Immune-cell populations, signal transduction on activation, and effector functions were studied. RESULTS: The patients had hypogammaglobulinemia or agammaglobulinemia, and their peripheral-blood B cells and T cells were almost exclusively of naive phenotype. Regulatory T cells and γδ T cells were absent. All patients carried a homozygous duplication--c.1292dupG in exon 13 of IKBKB, which encodes IκB kinase 2 (IKK2, also known as IKKß)--leading to loss of expression of IKK2, a component of the IKK-nuclear factor κB (NF-κB) pathway. Immune cells from the patients had impaired responses to stimulation through T-cell receptors, B-cell receptors, toll-like receptors, inflammatory cytokine receptors, and mitogens. CONCLUSIONS: A form of human SCID is characterized by normal lymphocyte development despite a loss of IKK2 function. IKK2 deficiency results in an impaired response to activation stimuli in a variety of immune cells, leading to clinically relevant impairment of adaptive and innate immunity. Although Ikk2 deficiency is lethal in mouse embryos, our observations suggest a more restricted, unique role of IKK2-NF-κB signaling in humans. (Funded by the German Federal Ministry of Education and Research and others.).


Asunto(s)
Agammaglobulinemia/genética , Quinasa I-kappa B/genética , Mutación , Inmunodeficiencia Combinada Grave/genética , Inmunidad Adaptativa/genética , Linfocitos B/fisiología , Resultado Fatal , Femenino , Genes Recesivos , Humanos , Quinasa I-kappa B/deficiencia , Inmunidad Innata/genética , Indígenas Norteamericanos , Lactante , Recién Nacido , Activación de Linfocitos , Recuento de Linfocitos , Masculino , Linaje , Análisis de Secuencia de ADN , Linfocitos T/fisiología
20.
Blood ; 124(6): 851-60, 2014 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-24894771

RESUMEN

Accumulation of CD3(+) T-cell receptor (TCR)αß(+)CD4(-)CD8(-) double-negative T cells (DNT) is a hallmark of autoimmune lymphoproliferative syndrome (ALPS). DNT origin and differentiation pathways remain controversial. Here we show that human ALPS DNT have features of terminally differentiated effector memory T cells reexpressing CD45RA(+) (TEMRA), but are CD27(+)CD28(+)KLRG1(-) and do not express the transcription factor T-bet. This unique phenotype was also detected among CD4(+) or CD8(+) ALPS TEMRA cells. T-cell receptor ß deep sequencing revealed a significant fraction of shared CDR3 sequences between ALPS DNT and both CD4(+) and CD8(+)TEMRA cells. Moreover, in ALPS patients with a germ line FAS mutation and somatic loss of heterozygosity, in whom biallelic mutant cells can be tracked by absent Fas expression, Fas-negative T cells accumulated not only among DNT, but also among CD4(+) and CD8(+)TEMRA cells. These data indicate that in human Fas deficiency DNT cannot only derive from CD8(+), but also from CD4(+) T cells. Furthermore, defective Fas signaling leads to aberrant transcriptional programs and differentiation of subsets of CD4(+) and CD8(+) T cells. Accumulation of these cells before their double-negative state appears to be an important early event in the pathogenesis of lymphoproliferation in ALPS patients.


Asunto(s)
Síndrome Linfoproliferativo Autoinmune/inmunología , Síndrome Linfoproliferativo Autoinmune/patología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/patología , Receptor fas/deficiencia , Receptor fas/genética , Adolescente , Adulto , Síndrome Linfoproliferativo Autoinmune/genética , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Niño , Preescolar , Estudios de Asociación Genética , Mutación de Línea Germinal , Humanos , Memoria Inmunológica , Antígenos Comunes de Leucocito/metabolismo , Pérdida de Heterocigocidad , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Proteínas de Dominio T Box/metabolismo , Subgrupos de Linfocitos T/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA