Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Immunol ; 41: 561-585, 2023 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-37126418

RESUMEN

Infection with SARS-CoV-2 results in clinical outcomes ranging from silent or benign infection in most individuals to critical pneumonia and death in a few. Genetic studies in patients have established that critical cases can result from inborn errors of TLR3- or TLR7-dependent type I interferon immunity, or from preexisting autoantibodies neutralizing primarily IFN-α and/or IFN-ω. These findings are consistent with virological studies showing that multiple SARS-CoV-2 proteins interfere with pathways of induction of, or response to, type I interferons. They are also congruent with cellular studies and mouse models that found that type I interferons can limit SARS-CoV-2 replication in vitro and in vivo, while their absence or diminution unleashes viral growth. Collectively, these findings point to insufficient type I interferon during the first days of infection as a general mechanism underlying critical COVID-19 pneumonia, with implications for treatment and directions for future research.


Asunto(s)
COVID-19 , Interferón Tipo I , Ratones , Humanos , Animales , Interferones/farmacología , SARS-CoV-2
2.
Nat Immunol ; 21(12): 1528-1539, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33020661

RESUMEN

Mutations that impact immune cell migration and result in immune deficiency illustrate the importance of cell movement in host defense. In humans, loss-of-function mutations in DOCK8, a guanine exchange factor involved in hematopoietic cell migration, lead to immunodeficiency and, paradoxically, allergic disease. Here, we demonstrate that, like humans, Dock8-/- mice have a profound type 2 CD4+ helper T (TH2) cell bias upon pulmonary infection with Cryptococcus neoformans and other non-TH2 stimuli. We found that recruited Dock8-/-CX3CR1+ mononuclear phagocytes are exquisitely sensitive to migration-induced cell shattering, releasing interleukin (IL)-1ß that drives granulocyte-macrophage colony-stimulating factor (GM-CSF) production by CD4+ T cells. Blocking IL-1ß, GM-CSF or caspase activation eliminated the type-2 skew in mice lacking Dock8. Notably, treatment of infected wild-type mice with apoptotic cells significantly increased GM-CSF production and TH2 cell differentiation. This reveals an important role for cell death in driving type 2 signals during infection, which may have implications for understanding the etiology of type 2 CD4+ T cell responses in allergic disease.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/deficiencia , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Células Th2/inmunología , Células Th2/metabolismo , Animales , Biomarcadores , Caspasas/metabolismo , Movimiento Celular/genética , Movimiento Celular/inmunología , Citocinas/genética , Citocinas/metabolismo , Susceptibilidad a Enfermedades , Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Inmunofenotipificación , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Ratones , Ratones Noqueados , Células Mieloides/inmunología , Células Mieloides/metabolismo , Fagocitos/inmunología , Fagocitos/metabolismo , Transducción de Señal
3.
Cell ; 159(7): 1578-90, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25525876

RESUMEN

Proteasomes and lysosomes constitute the major cellular systems that catabolize proteins to recycle free amino acids for energy and new protein synthesis. Tripeptidyl peptidase II (TPPII) is a large cytosolic proteolytic complex that functions in tandem with the proteasome-ubiquitin protein degradation pathway. We found that autosomal recessive TPP2 mutations cause recurrent infections, autoimmunity, and neurodevelopmental delay in humans. We show that a major function of TPPII in mammalian cells is to maintain amino acid levels and that TPPII-deficient cells compensate by increasing lysosome number and proteolytic activity. However, the overabundant lysosomes derange cellular metabolism by consuming the key glycolytic enzyme hexokinase-2 through chaperone-mediated autophagy. This reduces glycolysis and impairs the production of effector cytokines, including IFN-γ and IL-1ß. Thus, TPPII controls the balance between intracellular amino acid availability, lysosome number, and glycolysis, which is vital for adaptive and innate immunity and neurodevelopmental health.


Asunto(s)
Inmunidad Adaptativa , Aminopeptidasas/metabolismo , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Glucólisis , Inmunidad Innata , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/metabolismo , Proteolisis , Serina Endopeptidasas/metabolismo , Secuencia de Aminoácidos , Aminopeptidasas/química , Animales , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/química , Femenino , Humanos , Síndromes de Inmunodeficiencia/inmunología , Lisosomas/metabolismo , Masculino , Modelos Moleculares , Datos de Secuencia Molecular , Linaje , Alineación de Secuencia , Serina Endopeptidasas/química
4.
Immunol Rev ; 287(1): 9-19, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30565250

RESUMEN

DOCK8 immunodeficiency syndrome (DIDS) is a progressive combined immunodeficiency that can be distinguished from other combined immunodeficiencies or hyperimmunoglobulinemia E syndromes in featuring (a) profound susceptibility to virus infections of the skin, with associated skin cancers, and (b) severe food allergies. The DOCK8 locus has many repetitive sequence elements that predispose to the generation of large germline deletions as well as recombination-mediated somatic DNA repair. Residual DOCK8 protein contributes to the variable disease phenotype. The severe virus infections of the skin, and probably also VZV-associated vasculopathy, reflect an important function of DOCK8, which is normally required to maintain lymphocyte shape integrity as the cells migrate through dense tissues. Loss of DOCK8 also causes immune deficits through other mechanisms including a milder generalized cell survival defect and skewing of T helper cell subsets. Recent work has uncovered the roles for DOCK8 in dendritic cell responses that can also help explain the virus susceptibility, as well as in regulatory T cells that might help explain autoimmunity in a minority of patients. Fortunately, hematopoietic stem cell transplantation cures the eczema and infection susceptibility of DIDS, but not necessarily the other disease manifestations including food allergies.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/genética , Síndromes de Inmunodeficiencia/inmunología , Linfocitos T/inmunología , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Susceptibilidad a Enfermedades , Eccema , Terapia Genética , Trasplante de Células Madre Hematopoyéticas , Humanos , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/terapia , Neoplasias de Células Escamosas , Neumonía , Vasculitis del Sistema Nervioso Central
5.
J Clin Immunol ; 42(1): 119-129, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34657245

RESUMEN

Rare, biallelic loss-of-function mutations in DOCK8 result in a combined immune deficiency characterized by severe and recurrent cutaneous infections, eczema, allergies, and susceptibility to malignancy, as well as impaired humoral and cellular immunity and hyper-IgE. The advent of next-generation sequencing technologies has enabled the rapid molecular diagnosis of rare monogenic diseases, including inborn errors of immunity. These advances have resulted in the implementation of gene-guided treatments, such as hematopoietic stem cell transplant for DOCK8 deficiency. However, putative disease-causing variants revealed by next-generation sequencing need rigorous validation to demonstrate pathogenicity. Here, we report the eventual diagnosis of DOCK8 deficiency in a consanguineous family due to a novel homozygous intronic deletion variant that caused aberrant exon splicing and subsequent loss of expression of DOCK8 protein. Remarkably, the causative variant was not initially detected by clinical whole-genome sequencing but was subsequently identified and validated by combining advanced genomic analysis, RNA-seq, and flow cytometry. This case highlights the need to adopt multipronged confirmatory approaches to definitively solve complex genetic cases that result from variants outside protein-coding exons and conventional splice sites.


Asunto(s)
Síndrome de Job , Consanguinidad , Factores de Intercambio de Guanina Nucleótido/genética , Homocigoto , Humanos , Síndrome de Job/diagnóstico , Síndrome de Job/genética , Mutación/genética , Secuenciación del Exoma
6.
Hum Genet ; 140(9): 1299-1312, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34185153

RESUMEN

Genetic defects of innate immunity impairing intestinal bacterial sensing are linked to the development of Inflammatory Bowel Disease (IBD). Although much evidence supports a role of the intestinal virome in gut homeostasis, most studies focus on intestinal viral composition rather than on host intestinal viral sensitivity. To demonstrate the association between the development of Very Early Onset IBD (VEOIBD) and variants in the IFIH1 gene which encodes MDA5, a key cytosolic sensor for viral nucleic acids. Whole exome sequencing (WES) was performed in two independent cohorts of children with VEOIBD enrolled in Italy (n = 18) and USA (n = 24). Luciferase reporter assays were employed to assess MDA5 activity. An enrichment analysis was performed on IFIH1 comparing 42 VEOIBD probands with 1527 unrelated individuals without gastrointestinal or immunological issues. We identified rare, likely loss-of-function (LoF), IFIH1 variants in eight patients with VEOIBD from a combined cohort of 42 children. One subject, carrying a homozygous truncating variant resulting in complete LoF, experienced neonatal-onset, pan-gastrointestinal, IBD-like enteropathy plus multiple infectious episodes. The remaining seven subjects, affected by VEOIBD without immunodeficiency, were carriers of one LoF variant in IFIH1. Among these, two patients also carried a second hypomorphic variant, with partial function apparent when MDA5 was weakly stimulated. Furthermore, IFIH1 variants were significantly enriched in children with VEOIBD as compared to controls (p = 0.007). Complete and partial MDA5 deficiency is associated with VEOIBD with variable penetrance and expressivity, suggesting a role for impaired intestinal viral sensing in IBD pathogenesis.


Asunto(s)
Enfermedades Inflamatorias del Intestino/genética , Helicasa Inducida por Interferón IFIH1/genética , Mutación con Pérdida de Función , Preescolar , Femenino , Humanos , Lactante , Italia , Masculino , Secuenciación Completa del Genoma
7.
J Biol Chem ; 294(37): 13638-13656, 2019 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-31337704

RESUMEN

Magnesium transporter 1 (MAGT1) critically mediates magnesium homeostasis in eukaryotes and is highly-conserved across different evolutionary branches. In humans, loss-of-function mutations in the MAGT1 gene cause X-linked magnesium deficiency with Epstein-Barr virus (EBV) infection and neoplasia (XMEN), a disease that has a broad range of clinical and immunological consequences. We have previously shown that EBV susceptibility in XMEN is associated with defective expression of the antiviral natural-killer group 2 member D (NKG2D) protein and abnormal Mg2+ transport. New evidence suggests that MAGT1 is the human homolog of the yeast OST3/OST6 proteins that form an integral part of the N-linked glycosylation complex, although the exact contributions of these perturbations in the glycosylation pathway to disease pathogenesis are still unknown. Using MS-based glycoproteomics, along with CRISPR/Cas9-KO cell lines, natural killer cell-killing assays, and RNA-Seq experiments, we now demonstrate that humans lacking functional MAGT1 have a selective deficiency in both immune and nonimmune glycoproteins, and we identified several critical glycosylation defects in important immune-response proteins and in the expression of genes involved in immunity, particularly CD28. We show that MAGT1 function is partly interchangeable with that of the paralog protein tumor-suppressor candidate 3 (TUSC3) but that each protein has a different tissue distribution in humans. We observed that MAGT1-dependent glycosylation is sensitive to Mg2+ levels and that reduced Mg2+ impairs immune-cell function via the loss of specific glycoproteins. Our findings reveal that defects in protein glycosylation and gene expression underlie immune defects in an inherited disease due to MAGT1 deficiency.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Deficiencia de Magnesio/genética , Neoplasias/genética , Proteínas de Transporte de Catión/genética , Infecciones por Virus de Epstein-Barr/genética , Glicoproteínas/metabolismo , Glicosilación , Células HEK293 , Homeostasis , Humanos , Células Asesinas Naturales/metabolismo , Magnesio/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mutación , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
8.
Curr Opin Pediatr ; 31(6): 815-820, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31693592

RESUMEN

PURPOSE OF REVIEW: Studying primary immunodeficiencies (PIDs) provides insights into human antiviral immunity in the natural infectious environment. This review describes new PIDs with genetic defects that impair innate antiviral responses. RECENT FINDINGS: New genetic defects in the interferon (IFN) signaling pathway include IFNAR1 deficiency, which causes uncontrolled infections with measles-mumps-rubella or yellow fever vaccines, and possibly also cytomegalovirus (CMV); and IRF9 deficiency, which results in influenza virus susceptibility. Genetic defects in several pattern recognition receptors include MDA5 deficiency, which impairs viral RNA sensing and confers human rhinovirus susceptibility; RNA polymerase III haploinsufficiency, which impairs sensing of A:T-rich virus DNA and confers VZV susceptibility; and TLR3 deficiency, which causes HSV-1 encephalitis (HSE) or influenza virus pneumonitis. Defects in RNA metabolism, such as that caused by Debranching enzyme 1 deficiency, can cause virus meningoencephalitis. Finally, defects in host restriction factors for virus replication, such as in CIB1 deficiency, contribute to uncontrolled ß-HPV infections. SUMMARY: Several new PIDs highlight the role of type I/III IFN signaling pathway, virus sensors, and host virus restriction factors in human antiviral immunity.


Asunto(s)
Antivirales/uso terapéutico , Inmunidad Innata/efectos de los fármacos , Gripe Humana , Interferón Tipo I/genética , Enfermedades de Inmunodeficiencia Primaria/tratamiento farmacológico , Humanos , Interferón Tipo I/deficiencia , Transducción de Señal
9.
J Clin Immunol ; 36(5): 441-9, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27207373

RESUMEN

Since the discovery of the genetic basis of DOCK8 immunodeficiency syndrome (DIDS) in 2009, several hundred patients worldwide have been reported, validating and extending the initial clinical descriptions. Importantly, the beneficial role of hematopoietic stem cell transplantation for this disease has emerged, providing impetus for improved diagnosis. Additionally, several groups have further elucidated the biological functions of DOCK8 in the immune system that help explain disease pathogenesis. Here, we summarize these recent developments.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Factores de Intercambio de Guanina Nucleótido/genética , Trasplante de Células Madre Hematopoyéticas , Síndromes de Inmunodeficiencia/inmunología , Células T Asesinas Naturales/inmunología , Animales , Humanos , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/terapia , Memoria Inmunológica , Linfopenia , Mutación/genética
10.
J Allergy Clin Immunol ; 133(6): 1667-75, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24797421

RESUMEN

BACKGROUND: Autosomal recessive loss-of-function mutations in dedicator of cytokinesis 8 (DOCK8) cause a combined immunodeficiency characterized by atopy, recurrent infections, and cancer susceptibility. A genotype-phenotype explanation for the variable disease expression is lacking. OBJECTIVE: We investigated whether reversions contributed to the variable disease expression. METHODS: Patients followed at the National Institutes of Health's Clinical Center were studied. We performed detailed genetic analyses and intracellular flow cytometry to detect DOCK8 protein expression within lymphocyte subsets. RESULTS: We identified 17 of 34 DOCK8-deficient patients who had germline mutations with variable degrees of reversion caused by somatic repair. Somatic repair of the DOCK8 mutations resulted from second-site mutation, original-site mutation, gene conversion, and intragenic crossover. Higher degrees of reversion were associated with recombination-mediated repair. DOCK8 expression was restored primarily within antigen-experienced T cells or natural killer cells but less so in naive T or B cells. Several patients exhibited multiple different repair events. Patients who had reversions were older and had less severe allergic disease, although infection susceptibility persisted. No patients were cured without hematopoietic cell transplantation. CONCLUSIONS: In patients with DOCK8 deficiency, only certain combinations of germline mutations supported secondary somatic repair. Those patients had an ameliorated disease course with longer survival but still had fatal complications or required hematopoietic cell transplantation. These observations support the concept that some DOCK8-immunodeficient patients have mutable mosaic genomes that can modulate disease phenotype over time.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/genética , Síndromes de Inmunodeficiencia/genética , Mutación , Fenotipo , Adolescente , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Niño , Preescolar , Análisis Mutacional de ADN , Reparación del ADN , Genotipo , Mutación de Línea Germinal , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Síndromes de Inmunodeficiencia/diagnóstico , Síndromes de Inmunodeficiencia/mortalidad , Inmunofenotipificación , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo
11.
J Allergy Clin Immunol ; 133(5): 1400-9, 1409.e1-5, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24589341

RESUMEN

BACKGROUND: Identifying genetic syndromes that lead to significant atopic disease can open new pathways for investigation and intervention in allergy. OBJECTIVE: We sought to define a genetic syndrome of severe atopy, increased serum IgE levels, immune deficiency, autoimmunity, and motor and neurocognitive impairment. METHODS: Eight patients from 2 families with similar syndromic features were studied. Thorough clinical evaluations, including brain magnetic resonance imaging and sensory evoked potentials, were performed. Peripheral lymphocyte flow cytometry, antibody responses, and T-cell cytokine production were measured. Whole-exome sequencing was performed to identify disease-causing mutations. Immunoblotting, quantitative RT-PCR, enzymatic assays, nucleotide sugar, and sugar phosphate analyses, along with matrix-assisted laser desorption/ionization-time-of-flight mass spectrometry of glycans, were used to determine the molecular consequences of the mutations. RESULTS: Marked atopy and autoimmunity were associated with increased T(H)2 and T(H)17 cytokine production by CD4(+) T cells. Bacterial and viral infection susceptibility were noted along with T-cell lymphopenia, particularly of CD8(+) T cells, and reduced memory B-cell numbers. Apparent brain hypomyelination resulted in markedly delayed evoked potentials and likely contributed to neurologic abnormalities. Disease segregated with novel autosomal recessive mutations in a single gene, phosphoglucomutase 3 (PGM3). Although PGM3 protein expression was variably diminished, impaired function was demonstrated by decreased enzyme activity and reduced uridine diphosphate-N-acetyl-D-glucosamine, along with decreased O- and N-linked protein glycosylation in patients' cells. These results define a new congenital disorder of glycosylation. CONCLUSIONS: Autosomal recessive hypomorphic PGM3 mutations underlie a disorder of severe atopy, immune deficiency, autoimmunity, intellectual disability, and hypomyelination.


Asunto(s)
Enfermedades Autoinmunes/genética , Trastornos del Conocimiento/genética , Inmunodeficiencia Variable Común/genética , Enfermedades Genéticas Congénitas/genética , Hipersensibilidad/genética , Mutación , Fosfoglucomutasa/genética , Enfermedades Autoinmunes/enzimología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Linfocitos B/enzimología , Linfocitos B/inmunología , Linfocitos B/patología , Linfocitos T CD8-positivos/enzimología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Niño , Preescolar , Trastornos del Conocimiento/enzimología , Trastornos del Conocimiento/inmunología , Trastornos del Conocimiento/patología , Inmunodeficiencia Variable Común/enzimología , Inmunodeficiencia Variable Común/inmunología , Inmunodeficiencia Variable Común/patología , Familia , Femenino , Enfermedades Genéticas Congénitas/enzimología , Enfermedades Genéticas Congénitas/inmunología , Enfermedades Genéticas Congénitas/patología , Humanos , Hipersensibilidad/enzimología , Hipersensibilidad/inmunología , Hipersensibilidad/patología , Inmunoglobulina E/genética , Inmunoglobulina E/inmunología , Masculino , Linaje , Fosfoglucomutasa/inmunología , Fosfoglucomutasa/metabolismo , Células Th17/enzimología , Células Th17/inmunología , Células Th17/patología , Células Th2/enzimología , Células Th2/inmunología , Células Th2/patología , Adulto Joven
12.
J Biol Chem ; 286(45): 38913-23, 2011 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-21940623

RESUMEN

Dendritic Cells (DCs) play an important role in the initiation of the immune response by migrating to regional lymph nodes and presenting antigen processed at the inflammatory site to antigen-specific naïve T cells. Prostaglandin E2 (PGE2) has been reported to play an essential role in DC migration. We reported previously that PGE2 induces matrix metalloproteinase 9 (MMP-9) expression in DCs and that PGE2-induced MMP-9 is required for DC migration in vivo and in vitro. In this study, we investigated the signaling mechanisms involved in PGE2-induced MMP-9 expression in DCs. We show that PGE2-induced MMP-9 expression is mediated primarily through the EP2/EP4 → cAMP → protein kinase A (PKA)/PI3K → ERK signaling pathway, leading to c-Fos expression, and through JNK-mediated activation of c-Jun in a PKA/PI3K/ERK-independent manner. EP2 and EP4 receptor agonists, as well as cAMP analogs, mimic the up-regulation of MMP-9 by PGE2. PKA, PI3K, and ERK inhibitors abolished PGE2- and cAMP-induced c-Fos and MMP-9 up-regulation, and ERK activation was required for the binding of activator protein 1 (AP-1) transcription factor to the MMP-9 promoter. Our results describe a new molecular mechanism for the effect of PGE2 on MMP-9 production in DCs that could lead to future therapeutic approaches using ERK inhibitors to regulate DC migration.


Asunto(s)
Células Dendríticas/metabolismo , Dinoprostona/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Metaloproteinasa 9 de la Matriz/biosíntesis , Factor de Transcripción AP-1/metabolismo , Regulación hacia Arriba/fisiología , Animales , AMP Cíclico/genética , AMP Cíclico/metabolismo , Células Dendríticas/citología , Dinoprostona/farmacología , Inducción Enzimática/efectos de los fármacos , Inducción Enzimática/fisiología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/genética , Ratones , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Factor de Transcripción AP-1/genética , Regulación hacia Arriba/efectos de los fármacos
13.
J Clin Immunol ; 32(4): 698-708, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22476911

RESUMEN

We describe seven Turkish children with DOCK8 deficiency who have not been previously reported. Three patients presented with typical features of recurrent or severe cutaneous viral infections, atopic dermatitis, and recurrent respiratory or gastrointestinal tract infections. However, four patients presented with other features. Patient 1-1 featured sclerosing cholangitis and colitis; patient 2-1, granulomatous soft tissue lesion and central nervous system involvement, with primary central nervous system lymphoma found on follow-up; patient 3-1, a fatal metastatic leiomyosarcoma; and patient 4-2 showed no other symptoms initially besides atopic dermatitis. Similar to other previously reported Turkish patients, but in contrast to patients of non-Turkish ethnicity, the patients' lymphopenia was primarily restricted to CD4(+) T cells. Patients had homozygous mutations in DOCK8 that altered splicing, introduced premature terminations, destabilized protein, or involved large deletions within the gene. Genotyping of remaining family members showed that DOCK8 deficiency is a fully penetrant, autosomal recessive disease. In our patients, bone marrow transplantation resulted in rapid improvement followed by disappearance of viral skin lesions, including lesions resembling epidermodysplasia verruciformis, atopic dermatitis, and recurrent infections. Particularly for patients who feature unusual clinical manifestations, immunological testing, in conjunction with genetic testing, can prove invaluable in diagnosing DOCK8 deficiency and providing potentially curative treatment.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Factores de Intercambio de Guanina Nucleótido/deficiencia , Factores de Intercambio de Guanina Nucleótido/genética , Inmunodeficiencia Combinada Grave/genética , Adolescente , Trasplante de Médula Ósea , Niño , Preescolar , Dermatitis Atópica/genética , Femenino , Genotipo , Humanos , Inmunoglobulinas Intravenosas/uso terapéutico , Linfopenia/genética , Linfopenia/terapia , Masculino , Eliminación de Secuencia , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/terapia , Turquía
14.
N Engl J Med ; 361(21): 2046-55, 2009 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-19776401

RESUMEN

BACKGROUND: Recurrent sinopulmonary and cutaneous viral infections with elevated serum levels of IgE are features of some variants of combined immunodeficiency. The genetic causes of these variants are unknown. METHODS: We collected longitudinal clinical data on 11 patients from eight families who had recurrent sinopulmonary and cutaneous viral infections. We performed comparative genomic hybridization arrays and targeted gene sequencing. Variants with predicted loss-of-expression mutations were confirmed by means of a quantitative reverse-transcriptase-polymerase-chain-reaction assay and immunoblotting. We evaluated the number and function of lymphocytes with the use of in vitro assays and flow cytometry. RESULTS: Patients had recurrent otitis media, sinusitis, and pneumonias; recurrent Staphylococcus aureus skin infections with otitis externa; recurrent, severe herpes simplex virus or herpes zoster infections; extensive and persistent infections with molluscum contagiosum; and human papillomavirus infections. Most patients had severe atopy with anaphylaxis; several had squamous-cell carcinomas, and one had T-cell lymphoma-leukemia. Elevated serum IgE levels, hypereosinophilia, low numbers of T cells and B cells, low serum IgM levels, and variable IgG antibody responses were common. Expansion in vitro of activated CD8 T cells was impaired. Novel homozygous or compound heterozygous deletions and point mutations in the gene encoding the dedicator of cytokinesis 8 protein (DOCK8) led to the absence of DOCK8 protein in lymphocytes. CONCLUSIONS: Autosomal recessive DOCK8 deficiency is associated with a novel variant of combined immunodeficiency.


Asunto(s)
Eliminación de Gen , Factores de Intercambio de Guanina Nucleótido/genética , Inmunodeficiencia Combinada Grave/genética , Linfocitos T CD8-positivos/fisiología , Femenino , Genes Recesivos , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Inmunoglobulinas/sangre , Estudios Longitudinales , Activación de Linfocitos , Masculino , Linaje , Infecciones del Sistema Respiratorio/genética , Infecciones del Sistema Respiratorio/inmunología , Inmunodeficiencia Combinada Grave/inmunología , Enfermedades Cutáneas Infecciosas/genética , Enfermedades Cutáneas Infecciosas/inmunología
15.
J Exp Med ; 219(8)2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35708626

RESUMEN

Recessive or dominant inborn errors of type I interferon (IFN) immunity can underlie critical COVID-19 pneumonia in unvaccinated adults. The risk of COVID-19 pneumonia in unvaccinated children, which is much lower than in unvaccinated adults, remains unexplained. In an international cohort of 112 children (<16 yr old) hospitalized for COVID-19 pneumonia, we report 12 children (10.7%) aged 1.5-13 yr with critical (7 children), severe (3), and moderate (2) pneumonia and 4 of the 15 known clinically recessive and biochemically complete inborn errors of type I IFN immunity: X-linked recessive TLR7 deficiency (7 children) and autosomal recessive IFNAR1 (1), STAT2 (1), or TYK2 (3) deficiencies. Fibroblasts deficient for IFNAR1, STAT2, or TYK2 are highly vulnerable to SARS-CoV-2. These 15 deficiencies were not found in 1,224 children and adults with benign SARS-CoV-2 infection without pneumonia (P = 1.2 × 10-11) and with overlapping age, sex, consanguinity, and ethnicity characteristics. Recessive complete deficiencies of type I IFN immunity may underlie ∼10% of hospitalizations for COVID-19 pneumonia in children.


Asunto(s)
COVID-19 , Interferón Tipo I , Neumonía , Adulto , COVID-19/genética , Niño , Humanos , Patrón de Herencia , SARS-CoV-2
16.
J Exp Med ; 218(12)2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34726731

RESUMEN

Enterovirus (EV) infection rarely results in life-threatening infection of the central nervous system. We report two unrelated children with EV30 and EV71 rhombencephalitis. One patient carries compound heterozygous TLR3 variants (loss-of-function F322fs2* and hypomorphic D280N), and the other is homozygous for an IFIH1 variant (loss-of-function c.1641+1G>C). Their fibroblasts respond poorly to extracellular (TLR3) or intracellular (MDA5) poly(I:C) stimulation. The baseline (TLR3) and EV-responsive (MDA5) levels of IFN-ß in the patients' fibroblasts are low. EV growth is enhanced at early and late time points of infection in TLR3- and MDA5-deficient fibroblasts, respectively. Treatment with exogenous IFN-α2b before infection renders both cell lines resistant to EV30 and EV71, whereas post-infection treatment with IFN-α2b rescues viral susceptibility fully only in MDA5-deficient fibroblasts. Finally, the poly(I:C) and viral phenotypes of fibroblasts are rescued by the expression of WT TLR3 or MDA5. Human TLR3 and MDA5 are critical for cell-intrinsic immunity to EV, via the control of baseline and virus-induced type I IFN production, respectively.


Asunto(s)
Encefalitis Viral/inmunología , Infecciones por Enterovirus/inmunología , Helicasa Inducida por Interferón IFIH1/genética , Receptor Toll-Like 3/genética , Células Cultivadas , Preescolar , Encefalitis Viral/genética , Enterovirus/efectos de los fármacos , Enterovirus/fisiología , Infecciones por Enterovirus/genética , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/inmunología , Fibroblastos/virología , Humanos , Lactante , Interferón alfa-2/farmacología , Helicasa Inducida por Interferón IFIH1/inmunología , Interferón beta/inmunología , Interferón beta/metabolismo , Mutación con Pérdida de Función , Masculino , Errores Innatos del Metabolismo/genética , Errores Innatos del Metabolismo/inmunología , Poli I-C/farmacología , Rombencéfalo/virología , Receptor Toll-Like 3/inmunología , Replicación Viral/efectos de los fármacos
17.
Mol Cell Biol ; 27(12): 4551-65, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17438135

RESUMEN

GATA factors orchestrate hematopoiesis via multistep transcriptional mechanisms, but the interrelationships and importance of individual steps are poorly understood. Using complementation analysis with GATA-1-null cells and mice containing a hypomorphic allele of the chromatin remodeler BRG1, we dissected the pathway from GATA-1 binding to cofactor recruitment, chromatin loop formation, and transcriptional activation. Analysis of GATA-1-mediated activation of the beta-globin locus, in which GATA-1 assembles dispersed complexes at the promoters and the distal locus control region (LCR), revealed molecular intermediates, including GATA-1-independent and GATA-1-containing LCR subcomplexes, both defective in promoting loop formation. An additional intermediate consisted of an apparently normal LCR complex and a promoter complex with reduced levels of total RNA polymerase II (Pol II) and Pol II phosphorylated at serine 5 of the carboxy-terminal domain. Reduced BRG1 activity solely compromised Pol II and serine 5-phosphorylated Pol II occupancy at the promoter, phenocopying the LCR-deleted mouse. These studies defined a hierarchical order of GATA-1-triggered events at a complex locus and establish a novel mechanism of long-range gene regulation.


Asunto(s)
Diferenciación Celular , Cromatina/metabolismo , Factor de Transcripción GATA1/fisiología , Regulación de la Expresión Génica , Sistema Hematopoyético/fisiología , Alelos , Animales , Línea Celular , Cromatina/química , Inmunoprecipitación de Cromatina , ADN Helicasas/genética , ADN Helicasas/metabolismo , Células Eritroides/citología , Células Eritroides/metabolismo , Factor de Transcripción GATA1/genética , Eliminación de Gen , Globinas/genética , Región de Control de Posición , Ratones , Ratones Noqueados , Modelos Genéticos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína , ARN Polimerasa II/química , ARN Polimerasa II/metabolismo , Temperatura , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
Mol Cell Biol ; 26(19): 7056-67, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16980610

RESUMEN

The reciprocal expression of GATA-1 and GATA-2 during hematopoiesis is an important determinant of red blood cell development. Whereas Gata2 is preferentially transcribed early in hematopoiesis, elevated GATA-1 levels result in GATA-1 occupancy at sites upstream of the Gata2 locus and transcriptional repression. GATA-2 occupies these sites in the transcriptionally active locus, suggesting that a "GATA switch" abrogates GATA-2-mediated positive autoregulation. Chromatin immunoprecipitation (ChIP) coupled with genomic microarray analysis and quantitative ChIP analysis with GATA-1-null cells expressing an estrogen receptor ligand binding domain fusion to GATA-1 revealed additional GATA switches 77 kb upstream of Gata2 and within intron 4 at +9.5 kb. Despite indistinguishable GATA-1 occupancy at -77 kb and +9.5 kb versus other GATA switch sites, GATA-1 functioned uniquely at the different regions. GATA-1 induced histone deacetylation at and near Gata2 but not at the -77 kb region. The -77 kb region, which was DNase I hypersensitive in both active and inactive states, conferred equivalent enhancer activities in GATA-1- and GATA-2-expressing cells. By contrast, the +9.5 kb region exhibited considerably stronger enhancer activity in GATA-2- than in GATA-1-expressing cells, and other GATA switch sites were active only in GATA-1- or GATA-2-expressing cells. Chromosome conformation capture analysis demonstrated higher-order interactions between the -77 kb region and Gata2 in the active and repressed states. These results indicate that dispersed GATA factor complexes function via long-range chromatin interactions and qualitatively distinct activities to regulate Gata2 transcription.


Asunto(s)
Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/metabolismo , Factor de Transcripción GATA2/genética , Factor de Transcripción GATA2/metabolismo , Acetilación , Animales , Emparejamiento Base/genética , Secuencia de Bases , Cromatina/metabolismo , Inmunoprecipitación de Cromatina , Secuencia Conservada/genética , Desoxirribonucleasa I/metabolismo , Elementos de Facilitación Genéticos/genética , Humanos , Ratones , Datos de Secuencia Molecular , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas/genética , Receptores de Estrógenos/metabolismo , Factores de Transcripción p300-CBP/metabolismo
20.
J Exp Med ; 215(10): 2567-2585, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30143481

RESUMEN

Life-threatening pulmonary influenza can be caused by inborn errors of type I and III IFN immunity. We report a 5-yr-old child with severe pulmonary influenza at 2 yr. She is homozygous for a loss-of-function IRF9 allele. Her cells activate gamma-activated factor (GAF) STAT1 homodimers but not IFN-stimulated gene factor 3 (ISGF3) trimers (STAT1/STAT2/IRF9) in response to IFN-α2b. The transcriptome induced by IFN-α2b in the patient's cells is much narrower than that of control cells; however, induction of a subset of IFN-stimulated gene transcripts remains detectable. In vitro, the patient's cells do not control three respiratory viruses, influenza A virus (IAV), parainfluenza virus (PIV), and respiratory syncytial virus (RSV). These phenotypes are rescued by wild-type IRF9, whereas silencing IRF9 expression in control cells increases viral replication. However, the child has controlled various common viruses in vivo, including respiratory viruses other than IAV. Our findings show that human IRF9- and ISGF3-dependent type I and III IFN responsive pathways are essential for controlling IAV.


Asunto(s)
Alelos , Homocigoto , Gripe Humana , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/deficiencia , Orthomyxoviridae/inmunología , Neumonía Viral , Femenino , Humanos , Lactante , Gripe Humana/genética , Gripe Humana/inmunología , Gripe Humana/patología , Interferón alfa-2/genética , Interferón alfa-2/inmunología , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/inmunología , Neumonía Viral/genética , Neumonía Viral/inmunología , Neumonía Viral/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA