Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Blood Adv ; 3(2): 136-147, 2019 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-30651282

RESUMEN

Mutations in NCF1 (p47phox) cause autosomal recessive chronic granulomatous disease (CGD) with abnormal dihydrorhodamine (DHR) assay and absent p47phox protein. Genetic identification of NCF1 mutations is complicated by adjacent highly conserved (>98%) pseudogenes (NCF1B and NCF1C). NCF1 has GTGT at the start of exon 2, whereas the pseudogenes each delete 1 GT (ΔGT). In p47phox CGD, the most common mutation is ΔGT in NCF1 (c.75_76delGT; p.Tyr26fsX26). Sequence homology between NCF1 and its pseudogenes precludes reliable use of standard Sanger sequencing for NCF1 mutations and for confirming carrier status. We first established by flow cytometry that neutrophils from p47phox CGD patients had negligible p47phox expression, whereas those from p47phox CGD carriers had ∼60% of normal p47phox expression, independent of the specific mutation in NCF1 We developed a droplet digital polymerase chain reaction (ddPCR) with 2 distinct probes, recognizing either the wild-type GTGT sequence or the ΔGT sequence. A second ddPCR established copy number by comparison with the single-copy telomerase reverse transcriptase gene, TERT We showed that 84% of p47phox CGD patients were homozygous for ΔGT NCF1 The ddPCR assay also enabled determination of carrier status of relatives. Furthermore, only 79.2% of normal volunteers had 2 copies of GTGT per 6 total (NCF1/NCF1B/NCF1C) copies, designated 2/6; 14.7% had 3/6, and 1.6% had 4/6 GTGT copies. In summary, flow cytometry for p47phox expression quickly identifies patients and carriers of p47phox CGD, and genomic ddPCR identifies patients and carriers of ΔGT NCF1, the most common mutation in p47phox CGD.


Asunto(s)
Predisposición Genética a la Enfermedad , Enfermedad Granulomatosa Crónica/diagnóstico , Enfermedad Granulomatosa Crónica/genética , NADPH Oxidasas/deficiencia , Biomarcadores , Mapeo Cromosómico , Variaciones en el Número de Copia de ADN , Femenino , Citometría de Flujo , Expresión Génica , Estudios de Asociación Genética , Sitios Genéticos , Genotipo , Enfermedad Granulomatosa Crónica/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Linaje , Especies Reactivas de Oxígeno/metabolismo
2.
Curr Protoc Immunol ; 111: 7.23.1-7.23.16, 2015 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-26528633

RESUMEN

This unit describes the isolation of human polymorphonuclear neutrophils (PMN) from blood using dextran sedimentation and Percoll or Ficoll-Paque density gradients. Assays of neutrophil functions including respiratory burst activation, phagocytosis, and microbial killing are also described.


Asunto(s)
Centrifugación por Gradiente de Densidad/métodos , Citometría de Flujo/métodos , Neutrófilos/citología , Neutrófilos/inmunología , Citocromos c/metabolismo , Dextranos , Ficoll , Humanos , Peróxido de Hidrógeno/análisis , Peróxido de Hidrógeno/metabolismo , Fagocitosis/inmunología , Povidona , Estallido Respiratorio/fisiología , Dióxido de Silicio , Staphylococcus aureus/inmunología , Superóxido Dismutasa/metabolismo
3.
N Engl J Med ; 370(17): 1615-1625, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24716661

RESUMEN

Genetic defects in MOGS, the gene encoding mannosyl-oligosaccharide glucosidase (the first enzyme in the processing pathway of N-linked oligosaccharide), cause the rare congenital disorder of glycosylation type IIb (CDG-IIb), also known as MOGS-CDG. MOGS is expressed in the endoplasmic reticulum and is involved in the trimming of N-glycans. We evaluated two siblings with CDG-IIb who presented with multiple neurologic complications and a paradoxical immunologic phenotype characterized by severe hypogammaglobulinemia but limited clinical evidence of an infectious diathesis. A shortened immunoglobulin half-life was determined to be the mechanism underlying the hypogammaglobulinemia. Impaired viral replication and cellular entry may explain a decreased susceptibility to infections.


Asunto(s)
Agammaglobulinemia/genética , Trastornos Congénitos de Glicosilación/inmunología , Resistencia a la Enfermedad/genética , Virosis/inmunología , alfa-Glucosidasas/genética , Agammaglobulinemia/inmunología , Anticuerpos Antivirales/sangre , Niño , Trastornos Congénitos de Glicosilación/genética , Trastornos Congénitos de Glicosilación/metabolismo , Femenino , Glicosilación , Humanos , Inmunoglobulinas/metabolismo , Masculino
4.
Blood ; 123(15): 2308-16, 2014 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-24523241

RESUMEN

Warts, hypogammaglobulinemia, infections, and myelokathexis (WHIM) syndrome is a rare immunodeficiency disorder caused by gain-of-function mutations in the G protein-coupled chemokine receptor CXCR4. The CXCR4 antagonist plerixafor, which is approved by the US Food and Drug Administration (FDA) for stem cell mobilization in cancer and administered for that indication at 0.24 mg/kg, has been shown in short-term (1- to 2-week) phase 1 dose-escalation studies to correct neutropenia and other cytopenias in WHIM syndrome. However, long-term safety and long-term hematologic and clinical efficacy data are lacking. Here we report results from the first long-term clinical trial of plerixafor in any disease, in which 3 adults with WHIM syndrome self-injected 0.01 to 0.02 mg/kg (4% to 8% of the FDA-approved dose) subcutaneously twice daily for 6 months. Circulating leukocytes were durably increased throughout the trial in all patients, and this was associated with fewer infections and improvement in warts in combination with imiquimod; however, immunoglobulin levels and specific vaccine responses were not fully restored. No drug-associated side effects were observed. These results provide preliminary evidence for the safety and clinical efficacy of long-term, low-dose plerixafor in WHIM syndrome and support its continued study as mechanism-based therapy in this disease. The ClinicalTrials.gov identifier for this study is NCT00967785.


Asunto(s)
Compuestos Heterocíclicos/uso terapéutico , Síndromes de Inmunodeficiencia/tratamiento farmacológico , Receptores CXCR4/antagonistas & inhibidores , Verrugas/tratamiento farmacológico , Adulto , Bencilaminas , Ciclamas , Femenino , Citometría de Flujo , Compuestos Heterocíclicos/administración & dosificación , Compuestos Heterocíclicos/farmacocinética , Humanos , Masculino , Persona de Mediana Edad , Enfermedades de Inmunodeficiencia Primaria , Factores de Tiempo
5.
Blood ; 121(16): 3117-25, 2013 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-23430113

RESUMEN

Defective lymphocyte apoptosis results in chronic lymphadenopathy and/or splenomegaly associated with autoimmune phenomena. The prototype for human apoptosis disorders is the autoimmune lymphoproliferative syndrome (ALPS), which is caused by mutations in the FAS apoptotic pathway. Recently, patients with an ALPS-like disease called RAS-associated autoimmune leukoproliferative disorder, in which somatic mutations in NRAS or KRAS are found, also were described. Despite this progress, many patients with ALPS-like disease remain undefined genetically. We identified a homozygous, loss-of-function mutation in PRKCD (PKCδ) in a patient who presented with chronic lymphadenopathy, splenomegaly, autoantibodies, elevated immunoglobulins and natural killer dysfunction associated with chronic, low-grade Epstein-Barr virus infection. This mutation markedly decreased protein expression and resulted in ex vivo B-cell hyperproliferation, a phenotype similar to that of the PKCδ knockout mouse. Lymph nodes showed intense follicular hyperplasia, also mirroring the mouse model. Immunophenotyping of circulating lymphocytes demonstrated expansion of CD5+CD20+ B cells. Knockdown of PKCδ in normal mononuclear cells recapitulated the B-cell hyperproliferative phenotype in vitro. Reconstitution of PKCδ in patient-derived EBV-transformed B-cell lines partially restored phorbol-12-myristate-13-acetate-induced cell death. In summary, homozygous PRKCD mutation results in B-cell hyperproliferation and defective apoptosis with consequent lymphocyte accumulation and autoantibody production in humans, and disrupts natural killer cell function.


Asunto(s)
Síndrome Linfoproliferativo Autoinmune/genética , Linfocitos B/patología , Mutación , Proteína Quinasa C-delta/genética , Animales , Apoptosis , Síndrome Linfoproliferativo Autoinmune/complicaciones , Síndrome Linfoproliferativo Autoinmune/inmunología , Síndrome Linfoproliferativo Autoinmune/patología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Línea Celular , Proliferación Celular , Niño , Citocinas/inmunología , Infecciones por Virus de Epstein-Barr/complicaciones , Expresión Génica , Técnicas de Silenciamiento del Gen , Herpesvirus Humano 4/aislamiento & purificación , Homocigoto , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Enfermedades Linfáticas/complicaciones , Masculino , Ratones , Proteína Quinasa C-delta/inmunología , Esplenomegalia/complicaciones
6.
Blood ; 118(18): 4957-62, 2011 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-21890643

RESUMEN

WHIM syndrome is a rare congenital immunodeficiency disorder characterized by warts, hypogammaglobulinemia, infections, and myelokathexis (neutropenia because of impaired egress from the BM); most patients also have severe panleukopenia. Because WHIM syndrome is caused by mutations in the chemokine receptor CXCR4 that result in increased agonist-dependent signaling, we hypothesized that the CXCR4 antagonist plerixafor (Mozobil [Genyzme Corporation], AMD3100), might be an effective treatment. To test this, we enrolled 3 unrelated adult patients with the most common WHIM mutation, CXCR4(R334X), in a phase 1 dose-escalation study. Plerixafor increased absolute lymphocyte, monocyte, and neutrophil counts in blood to normal without significant side effects in all 3 patients. Peak responses occurred at 3-12 hours after injection and waned by 24 hours after injection which tracked the drug's pharmacokinetics. All 3 cell types increased in a dose-dependent manner with the rank order of responsiveness absolute lymphocyte > monocyte > neutrophil. These data provide the first pharmacologic evidence that panleukopenia in WHIM syndrome is caused by CXCL12-CXCR4 signaling-dependent leukocyte sequestration, and support continued study of plerixafor as mechanism-based therapy in this disease. This study is registered at http://www.clinicaltrials.gov as NCT00967785.


Asunto(s)
Compuestos Heterocíclicos/uso terapéutico , Síndromes de Inmunodeficiencia/tratamiento farmacológico , Linfopenia/tratamiento farmacológico , Verrugas/tratamiento farmacológico , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Fármacos Anti-VIH/administración & dosificación , Fármacos Anti-VIH/farmacocinética , Fármacos Anti-VIH/farmacología , Fármacos Anti-VIH/uso terapéutico , Bencilaminas , Recuento de Células Sanguíneas , Ciclamas , Relación Dosis-Respuesta a Droga , Femenino , Compuestos Heterocíclicos/administración & dosificación , Compuestos Heterocíclicos/farmacocinética , Compuestos Heterocíclicos/farmacología , Humanos , Síndromes de Inmunodeficiencia/sangre , Síndromes de Inmunodeficiencia/complicaciones , Síndromes de Inmunodeficiencia/genética , Leucocitos/efectos de los fármacos , Leucocitos/patología , Linfopenia/complicaciones , Linfopenia/patología , Masculino , Persona de Mediana Edad , Enfermedades de Inmunodeficiencia Primaria , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/genética , Resultado del Tratamiento , Verrugas/sangre , Verrugas/complicaciones , Verrugas/genética , Adulto Joven
7.
N Engl J Med ; 363(27): 2600-10, 2010 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-21190454

RESUMEN

BACKGROUND: Failure to generate phagocyte-derived superoxide and related reactive oxygen intermediates (ROIs) is the major defect in chronic granulomatous disease, causing recurrent infections and granulomatous complications. Chronic granulomatous disease is caused by missense, nonsense, frameshift, splice, or deletion mutations in the genes for p22(phox), p40(phox), p47(phox), p67(phox) (autosomal chronic granulomatous disease), or gp91(phox) (X-linked chronic granulomatous disease), which result in variable production of neutrophil-derived ROIs. We hypothesized that residual ROI production might be linked to survival in patients with chronic granulomatous disease. METHODS: We assessed the risks of illness and death among 287 patients with chronic granulomatous disease from 244 kindreds. Residual ROI production was measured with the use of superoxide-dependent ferricytochrome c reduction and flow cytometry with dihydrorhodamine oxidation assays. Expression of NADPH oxidase component protein was detected by means of immunoblotting, and the affected genes were sequenced to identify causal mutations. RESULTS: Survival of patients with chronic granulomatous disease was strongly associated with residual ROI production as a continuous variable, independently of the specific gene affected. Patients with mutations in p47(phox) and most missense mutations in gp91(phox) (with the exception of missense mutations in the nucleotide-binding and heme-binding domains) had more residual ROI production than patients with nonsense, frameshift, splice, or deletion mutations in gp91(phox). After adolescence, mortality curves diverged according to the extent of residual ROI production. CONCLUSIONS: Patients with chronic granulomatous disease and modest residual production of ROI have significantly less severe illness and a greater likelihood of long-term survival than patients with little residual ROI production. The production of residual ROI is predicted by the specific NADPH oxidase mutation, regardless of the specific gene affected, and it is a predictor of survival in patients with chronic granulomatous disease. (Funded by the National Institutes of Health.).


Asunto(s)
Enfermedad Granulomatosa Crónica/enzimología , NADPH Oxidasas/sangre , Especies Reactivas de Oxígeno/metabolismo , Análisis de Varianza , Femenino , Genotipo , Enfermedad Granulomatosa Crónica/sangre , Enfermedad Granulomatosa Crónica/genética , Enfermedad Granulomatosa Crónica/mortalidad , Humanos , Leucocitos Mononucleares/metabolismo , Masculino , Mutación , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Neutrófilos/metabolismo , Fenotipo , Modelos de Riesgos Proporcionales , Factores de Riesgo , Análisis de Secuencia de ADN , Índice de Severidad de la Enfermedad , Análisis de Supervivencia
8.
Blood ; 116(15): 2793-802, 2010 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-20616219

RESUMEN

Mutations in more than 15 genes are now known to cause severe congenital neutropenia (SCN); however, the pathologic mechanisms of most genetic defects are not fully defined. Deficiency of G6PC3, a glucose-6-phosphatase, causes a rare multisystem syndrome with SCN first described in 2009. We identified a family with 2 children with homozygous G6PC3 G260R mutations, a loss of enzymatic function, and typical syndrome features with the exception that their bone marrow biopsy pathology revealed abundant neutrophils consistent with myelokathexis. This pathologic finding is a hallmark of another type of SCN, WHIM syndrome, which is caused by gain-of-function mutations in CXCR4, a chemokine receptor and known neutrophil bone marrow retention factor. We found markedly increased CXCR4 expression on neutrophils from both our G6PC3-deficient patients and G6pc3(-/-) mice. In both patients, granulocyte colony-stimulating factor treatment normalized CXCR4 expression and neutrophil counts. In G6pc3(-/-) mice, the specific CXCR4 antagonist AMD3100 rapidly reversed neutropenia. Thus, myelokathexis associated with abnormally high neutrophil CXCR4 expression may contribute to neutropenia in G6PC3 deficiency and responds well to granulocyte colony-stimulating factor.


Asunto(s)
Glucosa-6-Fosfatasa/genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/genética , Neutropenia/congénito , Neutropenia/genética , Receptores CXCR4/genética , Adolescente , Animales , Niño , Femenino , Expresión Génica , Homocigoto , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación Missense , Neutropenia/enzimología , Neutrófilos/metabolismo , Síndrome
9.
Inflammation ; 30(5): 178-88, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17624583

RESUMEN

Fibrinogen, in addition to its role in coagulation, is also an acute phase protein of inflammation. Treatment of adherent human monocytes with fibrinogen increases IL-8, IL-6, and TNF-alpha, but has no effect on MCP-1, IFN-beta, or IP-10. Treatment of monocytes with fibrinogen and C5a doubles IL-8 and IL-6 production, compared to fibrinogen alone. The increase in cytokine production was accompanied by a transient increase in IL-8 mRNA and increased NF-kappaB activity. Monocytes from an IRAK-4- and two NEMO-deficient patients had 80% reduced IL-8 responses to fibrinogen. Moreover, responses to fibrinogen were blocked with anti-CD14 antibody (MY4), a subunit of the LPS receptor. The data indicate that fibrinogen alone and fibrinogen plus C5a are potent inducers of cytokine production in monocytes, and that signaling by fibrinogen is mediated through the TLR-4 pathway.


Asunto(s)
Fibrinógeno/metabolismo , Interleucina-8/metabolismo , Monocitos/metabolismo , Transducción de Señal , Receptor Toll-Like 4/metabolismo , Células Cultivadas , Complemento C5a/metabolismo , Relación Dosis-Respuesta a Droga , Fibrinógeno/farmacología , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Quinasas Asociadas a Receptores de Interleucina-1/genética , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Interleucina-6/metabolismo , Interleucina-8/genética , Receptores de Lipopolisacáridos/metabolismo , Monocitos/efectos de los fármacos , Monocitos/inmunología , Mutación , FN-kappa B/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...