Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Clin Immunol ; 36(4): 397-405, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27059040

RESUMEN

WHIM syndrome is an autosomal dominant immunodeficiency disease caused by mutations affecting the carboxy-terminus of CXCR4. To characterize novel genetic causes of the syndrome, we recruited a pediatric patient with possible WHIM syndrome, performed CXCR4 gene sequencing and compared his clinical phenotype and CXCR4 tail amino acid sequences with other patients with WHIM syndrome carrying CXCR4 (R334X) mutations. We identified and biochemically characterized a heterozygous 5 base pair deletion (nucleotides 986-990) located in the portion of the open reading frame (ORF) of CXCR4 that encodes the carboxy-terminal domain of the receptor. This CXCR4 (L329fs) mutation causes a frame-shift at codon 329 resulting in replacement of the final 24 predicted amino acids of the receptor with 12 missense amino acids. Like previously reported WHIM mutations, this frame-shift mutation CXCR4 (L329fs) decreased receptor downregulation in response to the CXCR4 agonist CXCL12 in patient PBMCs as well as in transfected K562 and HEK 293 cells, but increased calcium flux responses in K562 cells to CXCL12 stimulation. Thus, CXCR4 (L329fs) appears to be a de novo autosomal dominant frame-shift gain-of-function mutation that like other carboxy-terminus mutations causes WHIM syndrome. The same CXCR4 (L329fs) frame-shift variant has been reported to occur in tumor cells from a patient with Waldenström's Macroglobulemia (WM), but is caused by a distinct genetic mechanism: insertion of a single nucleotide in the L329 codon, providing additional evidence that the carboxy-terminus of CXCR4 is a genetic hotspot for mutation.


Asunto(s)
Síndromes de Inmunodeficiencia/genética , Receptores CXCR4/genética , Macroglobulinemia de Waldenström/genética , Verrugas/genética , Preescolar , Células HEK293 , Humanos , Células K562 , Masculino , Mutación , Neutropenia/genética , Enfermedades de Inmunodeficiencia Primaria
2.
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
3.
J Allergy Clin Immunol ; 133(6): 1676-85.e5, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24373354

RESUMEN

BACKGROUND: We previously reported abnormalities in circulating B cells in patients with chronic granulomatous disease (CGD) and those with HIV infection. Gastrointestinal complications are common to both diseases and likely involve perturbation of immune cells, including plasma cells (PCs). IgA is the most abundant immunoglobulin in the human body, with roles in protection and maintenance of intestinal homeostasis. IgA is produced primarily by PCs residing in mucosal tissues that are also thought to circulate in the blood. OBJECTIVE: We sought to characterize and compare PCs in patients with infectious (HIV) and noninfectious (CGD and Crohn disease) diseases that have been associated with intestinal inflammation. METHODS: Phenotypic and transcriptional analyses were performed on cells isolated from the blood and colon. RESULTS: IgA-secreting CCR10-expressing PCs predominated in the guts of healthy subjects, whereas in patients with HIV, CGD, and Crohn disease, there was a significant increase in the proportion of IgG-secreting PCs. Where intestinal inflammation was present, IgG-secreting PCs expressed reduced levels of CCR10 and increased levels of CXCR4. The intensity of CXCR4 expression correlated with the frequency of IgG-expressing PCs and the frequency of CXCR4(+)/IgG(+) PCs was associated with the severity of intestinal inflammatory disease yet distinct from PCs and plasmablasts circulating in the blood. CONCLUSIONS: These findings suggest that regardless of the underlying disease, the presence of CXCR4(+)/IgG(+) PCs in the gut is a strong yet localized indicator of intestinal inflammation. Furthermore, our findings suggest that CXCR4(+)/IgG(+) PCs might play a role in immune cell homeostasis during inflammatory processes of the gut.


Asunto(s)
Gastroenteritis/inmunología , Gastroenteritis/metabolismo , Inmunoglobulina G/metabolismo , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Receptores CXCR4/metabolismo , Adulto , Biopsia , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/metabolismo , Femenino , Gastroenteritis/genética , Enfermedad Granulomatosa Crónica/inmunología , Enfermedad Granulomatosa Crónica/metabolismo , Infecciones por VIH/inmunología , Infecciones por VIH/metabolismo , Humanos , Isotipos de Inmunoglobulinas/inmunología , Isotipos de Inmunoglobulinas/metabolismo , Inmunofenotipificación , Masculino , Persona de Mediana Edad , Membrana Mucosa/inmunología , Membrana Mucosa/metabolismo , Receptores Mensajeros de Linfocitos/genética , Receptores Mensajeros de Linfocitos/metabolismo , Adulto Joven
4.
Blood ; 120(1): 181-9, 2012 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-22596258

RESUMEN

WHIM syndrome is a rare, autosomal dominant, immunodeficiency disorder so-named because it is characterized by warts, hypogammaglobulinemia, infections, and myelokathexis (defective neutrophil egress from the BM). Gain-of-function mutations that truncate the C-terminus of the chemokine receptor CXCR4 by 10-19 amino acids cause WHIM syndrome. We have identified a family with autosomal dominant inheritance of WHIM syndrome that is caused by a missense mutation in CXCR4, E343K (1027G → A). This mutation is also located in the C-terminal domain, a region responsible for negative regulation of the receptor. Accordingly, like CXCR4(R334X), the most common truncation mutation in WHIM syndrome, CXCR4(E343K) mediated approximately 2-fold increased signaling in calcium flux and chemotaxis assays relative to wild-type CXCR4; however, CXCR4(E343K) had a reduced effect on blocking normal receptor down-regulation from the cell surface. Therefore, in addition to truncating mutations in the C-terminal domain of CXCR4, WHIM syndrome may be caused by a single charge-changing amino acid substitution in this domain, E343K, that results in increased receptor signaling.


Asunto(s)
Sustitución de Aminoácidos/genética , Síndromes de Inmunodeficiencia/genética , Receptores CXCR4/química , Receptores CXCR4/genética , Verrugas/genética , Secuencia de Aminoácidos , Niño , Preescolar , Salud de la Familia , Femenino , Humanos , Células K562 , Leucopenia/genética , Masculino , Datos de Secuencia Molecular , Linaje , Fenotipo , Enfermedades de Inmunodeficiencia Primaria , Estructura Terciaria de Proteína/genética
5.
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
6.
J Cell Mol Med ; 15(10): 2071-81, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21070597

RESUMEN

WHIM is an acronym for a rare immunodeficiency syndrome (OMIM #193670) caused by autosomal dominant mutations truncating the C-terminus of the chemokine receptor CXC chemokine receptor 4 (CXCR4). WHIM mutations may potentiate CXCR4 signalling, suggesting that the United States Food and Drug Administration (FDA)-approved CXCR4 antagonist AnorMED3100 (AMD3100) (also known as Plerixafor) may be beneficial in WHIM syndrome. We have tested this at the preclinical level by comparing Chinese hamster ovary (CHO) and K562 cell lines matched for expression of recombinant wild-type CXCR4 (CXCR4(WT)) and the most common WHIM variant of CXCR4 (CXCR4(R334X)), as well as leucocytes from a WHIM patient with the CXCR4(R334X) mutation versus healthy controls. We found that CXCR4(R334X) mediated modestly increased signalling (~2-fold) in all functional assays tested, but strongly resisted ligand-dependent down-regulation. AMD3100 was equipotent and equieffective as an antagonist at CXCR4(R334X) and CXCR4(WT) . Together, our data provide further evidence that CXCR4(R334X) is a gain-of-function mutation, and support clinical evaluation of AMD3100 as mechanism-based treatment in patients with WHIM syndrome.


Asunto(s)
Compuestos Heterocíclicos/farmacología , Síndromes de Inmunodeficiencia/genética , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/genética , Verrugas/genética , Adulto , Animales , Bencilaminas , Células CHO , Cricetinae , Ciclamas , Femenino , Citometría de Flujo , Regulación de la Expresión Génica , Humanos , Células K562 , Enfermedades de Inmunodeficiencia Primaria
7.
Am J Respir Crit Care Med ; 178(10): 1066-74, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18703788

RESUMEN

RATIONALE: Pulmonary nontuberculous mycobacterial (PNTM) disease is increasing, but predisposing features have been elusive. OBJECTIVES: To prospectively determine the morphotype, immunophenotype, and cystic fibrosis transmembrane conductance regulator genotype in a large cohort with PNTM. METHODS: We prospectively enrolled 63 patients with PNTM infection, each of whom had computerized tomography, echocardiogram, pulmonary function, and flow cytometry of peripheral blood. In vitro cytokine production in response to mitogen, LPS, and cytokines was performed. Anthropometric measurements were compared with National Health and Nutrition Examination Survey (NHANES) age- and ethnicity-matched female control subjects extracted from the NHANES 2001-2002 dataset. MEASUREMENTS AND MAIN RESULTS: Patients were 59.9 (+/-9.8 yr [SD]) old, and 5.4 (+/-7.9 yr) from diagnosis to enrollment. Patients were 95% female, 91% white, and 68% lifetime nonsmokers. A total of 46 were infected with Mycobacterium avium complex, M. xenopi, or M. kansasii; 17 were infected with rapidly growing mycobacteria. Female patients were significantly taller (164.7 vs. 161.0 cm; P < 0.001) and thinner (body mass index, 21.1 vs. 28.2; P < 0.001) than matched NHANES control subjects, and thinner (body mass index, 21.1 vs. 26.8; P = 0.002) than patients with disseminated nontuberculous mycobacterial infection. A total of 51% of patients had scoliosis, 11% pectus excavatum, and 9% mitral valve prolapse, all significantly more than reference populations. Stimulated cytokine production was similar to that of healthy control subjects, including the IFN-gamma/IL-12 pathway. CD4(+), CD8(+), B, and natural killer cell numbers were normal. A total of 36% of patients had mutations in the cystic fibrosis transmembrane conductance regulator gene. CONCLUSIONS: Patients with PNTM infection are taller and leaner than control subjects, with high rates of scoliosis, pectus excavatum, mitral valve prolapse, and cystic fibrosis transmembrane conductance regulator mutations, but without recognized immune defects.


Asunto(s)
Infecciones por Mycobacterium no Tuberculosas/etiología , Neumonía Bacteriana/etiología , Anciano , Estatura , Estudios de Casos y Controles , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Femenino , Tórax en Embudo/complicaciones , Humanos , Masculino , Persona de Mediana Edad , Mutación , Infecciones por Mycobacterium no Tuberculosas/genética , Infecciones por Mycobacterium no Tuberculosas/inmunología , Fenotipo , Estudios Prospectivos , Factores de Riesgo , Escoliosis/complicaciones , Factores Sexuales , Fumar/efectos adversos , Síndrome , Delgadez/complicaciones
8.
Sci Transl Med ; 8(335): 335ra57, 2016 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-27099176

RESUMEN

X-linked severe combined immunodeficiency (SCID-X1) is a profound deficiency of T, B, and natural killer (NK) cell immunity caused by mutations inIL2RGencoding the common chain (γc) of several interleukin receptors. Gamma-retroviral (γRV) gene therapy of SCID-X1 infants without conditioning restores T cell immunity without B or NK cell correction, but similar treatment fails in older SCID-X1 children. We used a lentiviral gene therapy approach to treat five SCID-X1 patients with persistent immune dysfunction despite haploidentical hematopoietic stem cell (HSC) transplant in infancy. Follow-up data from two older patients demonstrate that lentiviral vector γc transduced autologous HSC gene therapy after nonmyeloablative busulfan conditioning achieves selective expansion of gene-marked T, NK, and B cells, which is associated with sustained restoration of humoral responses to immunization and clinical improvement at 2 to 3 years after treatment. Similar gene marking levels have been achieved in three younger patients, albeit with only 6 to 9 months of follow-up. Lentiviral gene therapy with reduced-intensity conditioning appears safe and can restore humoral immune function to posthaploidentical transplant older patients with SCID-X1.


Asunto(s)
Terapia Genética/métodos , Células Madre Hematopoyéticas/metabolismo , Lentivirus/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia , Adolescente , Adulto , Linfocitos B/metabolismo , Niño , Vectores Genéticos/genética , Humanos , Subunidad gamma Común de Receptores de Interleucina/genética , Células Asesinas Naturales/metabolismo , Masculino , Linfocitos T/metabolismo , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Adulto Joven
9.
Pediatrics ; 114(2): 462-8, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15286231

RESUMEN

OBJECTIVE: Chronic granulomatous disease (CGD) is a rare disorder of phagocyte oxidative metabolism. In addition to infectious complications, granulomatous lesions often involve hollow viscera, especially the gastrointestinal (GI) tract. The objective of this study was to evaluate the clinical presentation, prevalence, and consequences of GI involvement in patients with CGD. METHODS: The medical records of 140 patients with CGD (67% X-linked) followed at the National Institutes of Health were reviewed and abstracted for GI manifestations. All available GI pathology was reviewed. RESULTS: GI involvement was recorded in 46 (32.8%) of 140 patients with CGD, 89% of whom had X-linked inheritance. The median age at the time of initial GI manifestations was 5 years (range: 0.8-30 years); 70% of the affected patients presented with GI involvement in the first decade of life. Abdominal pain was the most frequent symptom (100%), and hypoalbuminemia was the most frequent sign (70%). Prednisone controlled symptoms and signs in the majority of affected patients, but relapse of symptoms occurred in 71%. GI involvement had no effect on mortality and was unassociated with interferon-gamma use. CONCLUSION: GI involvement is a common and recurring problem in CGD, especially in those with X-linked inheritance. Currently, there is no clear evidence for an infectious cause. The frequency of GI involvement is unaffected by the use of interferon-gamma and does not affect mortality. GI involvement should be sought in patients who have CGD with abdominal pain, growth delay, or hypoalbuminemia.


Asunto(s)
Enfermedad Granulomatosa Crónica/complicaciones , Enfermedades Inflamatorias del Intestino/etiología , Dolor Abdominal/etiología , Adolescente , Adulto , Niño , Preescolar , Colon/patología , Estreñimiento/etiología , Diarrea/etiología , Femenino , Glucocorticoides/uso terapéutico , Enfermedad Granulomatosa Crónica/diagnóstico , Enfermedad Granulomatosa Crónica/tratamiento farmacológico , Enfermedad Granulomatosa Crónica/mortalidad , Trastornos del Crecimiento/etiología , Humanos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/patología , Masculino , Náusea/etiología , Prednisona/uso terapéutico , Prevalencia , Recurrencia , Estudios Retrospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA