Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Sci Rep ; 14(1): 13138, 2024 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-38849509

RESUMEN

Colorectal cancer (CRC) is a global health concern, and the incidence of early onset (EO) CRC, has an upward trend. This study delves into the genomic landscape of EO-CRC, specifically focusing on pediatric (PED) and young adult (YA) patients, comparing them with adult (AD) CRC. In this retrospective monocentric investigation, we performed targeted next-generation sequencing to compare the mutational profile of 38 EO-CRCs patients (eight PED and 30 YA) to those of a 'control group' consisting of 56 AD-CRCs. Our findings reveal distinct molecular profiles in EO-CRC, notably in the WNT and PI3K-AKT pathways. In pediatrics, we observed a significantly higher frequency of RNF43 mutations, whereas APC mutations were more prevalent in adult cases. These observations suggest age-related differences in the activation of the WNT pathway. Pathway and copy number variation analysis reveal that AD-CRC and YA-CRC have more similarities than the pediatric patients. PED shows a peculiar profile with CDK6 amplification and the enrichment of lysine degradation pathway. These findings may open doors for personalized therapies, such as PI3K-AKT pathway inhibitors or CDK6 inhibitors for pediatric patients. Additionally, the distinct molecular signatures of EO-CRC underscore the need for age-specific treatment strategies and precision medicine. This study emphasizes the importance of comprehensive molecular investigations in EO-CRCs, which can potentially improve diagnostic accuracy, prognosis, and therapeutic decisions for these patients. Collaboration between the pediatric and adult oncology community is fundamental to improve oncological outcomes for this rare and challenging pediatric tumor.


Asunto(s)
Neoplasias Colorrectales , Mutación , Humanos , Neoplasias Colorrectales/genética , Masculino , Femenino , Niño , Adulto Joven , Adolescente , Adulto , Estudios Retrospectivos , Preescolar , Variaciones en el Número de Copia de ADN , Genómica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Vía de Señalización Wnt/genética
2.
BMC Cancer ; 17(1): 107, 2017 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-28166781

RESUMEN

BACKGROUND: Desmoplastic small round cell tumor (DSRCT) is a rare and highly aggressive disease, that can be described as a member of the family of small round blue cell tumors. The molecular diagnostic marker is the t(11;22)(p13;q12) translocation, which creates an aberrant transcription factor, EWS-WT1, that underlies the oncogenesis of DSRCT. Current treatments are not very effective so new active drugs are needed. Trabectedin, now used as a single agent for the treatment of soft tissue sarcoma, was reported to be active in some pre-treated DSRCT patients. Using JN-DSRCT-1, a cell line derived from DSRCT expressing the EWS-WT1 fusion protein, we investigated the ability of trabectedin to modify the function of the chimeric protein, as in other sarcomas expressing fusion proteins. After detailed characterization of the EWS-WT1 transcripts structure, we investigated the mode of action of trabectedin, looking at the expression and function of the oncogenic chimera. METHODS: We characterized JN-DSRCT-1 cells using cellular approaches (FISH, Clonogenicity assay) and molecular approaches (Sanger sequencing, ChIP, GEP). RESULTS: JN-DSRCT-1 cells were sensitive to trabectedin at nanomolar concentrations. The cell line expresses different variants of EWS-WT1, some already identified in patients. EWS-WT1 mRNA expression was affected by trabectedin and chimeric protein binding on its target gene promoters was reduced. Expression profiling indicated that trabectedin affects the expression of genes involved in cell proliferation and apoptosis. CONCLUSIONS: The JN-DSRCT-1 cell line, in vitro, is sensitive to trabectedin: after drug exposure, EWS-WT1 chimera expression decreases as well as binding on its target promoters. Probably the heterogeneity of chimera transcripts is an obstacle to precisely defining the molecular mode of action of drugs, calling for further cellular models of DSRCT, possibly growing in vivo too, to mimic the biological complexity of this disease.


Asunto(s)
Tumor Desmoplásico de Células Pequeñas Redondas/tratamiento farmacológico , Dioxoles/farmacología , Regulación Neoplásica de la Expresión Génica , Proteínas de Fusión Oncogénica/efectos de los fármacos , Tetrahidroisoquinolinas/farmacología , Antineoplásicos Alquilantes/farmacología , Antineoplásicos Alquilantes/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Tumor Desmoplásico de Células Pequeñas Redondas/metabolismo , Tumor Desmoplásico de Células Pequeñas Redondas/fisiopatología , Dioxoles/uso terapéutico , Humanos , Proteínas de Fusión Oncogénica/genética , Proteína EWS de Unión a ARN , Tetrahidroisoquinolinas/uso terapéutico , Trabectedina , Proteínas WT1
3.
Clin Cancer Res ; 19(18): 5192-201, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23888069

RESUMEN

PURPOSE: To explore the value of triazines in solitary fibrous tumor (SFT). EXPERIMENTAL DESIGN: We retrospectively reviewed 8 cases of patients with SFT treated with dacarbazine (1,200 mg/m(2) every 3 weeks) as from January 2012. Then, we studied a dedifferentiated-SFT subcutaneously xenotransplanted into severe combined immunodeficient (SCID) mice. Dacarbazine, temozolomide, sunitinib, bevacizumab, and pazopanib were administered at their reported optimal doses for the mouse model when mean tumor volume (TV) was about 80 mm(3); each experimental groups included 6 mice. Drug activity was assessed as tumor volume inhibition percentage (TVI%). Dacarbazine was tested according to two different schedules of administration. One hunded twenty days after treatment interruption, mouse tumor samples were analyzed. RESULTS: Among the eight patients treated with dacarbazine, best response evaluation criteria in solid tumors responses (RECIST) were three partial responses, 4 stable disease, 1 progression. Two responsive patients had paraneoplastic hypoglycemia that disappeared after 10 days from starting dacarbazine. In the dedifferentiated-SFT xenograft model, dacarbazine and temozolomide showed the highest antitumor activity (about 95% TVI), confirmed pathologically. Sunitinib and pazopanib were only marginally active (52% and 41% TVI, respectively), whereas bevacizumab caused a 78% TVI. No tumor regrowth was observed up to 100 days from end of treatment with temozolomide and dacarbazine, whereas secondary progression followed sunitinib, pazopanib, and bevacizumab interruption. CONCLUSIONS: Dacarbazine as single agent has antitumor activity in SFT. Our preclinical results suggest a cytotoxic effect of temozolomide and dacarbazine, as compared with a cytostatic role for sunitinib, pazopanib, and bevacizumab. A phase II study on dacarbazine in advanced SFT is planned.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neovascularización Patológica/prevención & control , Tumores Fibrosos Solitarios/tratamiento farmacológico , Tumores Fibrosos Solitarios/patología , Adulto , Anciano , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Bevacizumab , Dacarbazina/administración & dosificación , Dacarbazina/análogos & derivados , Femenino , Estudios de Seguimiento , Humanos , Indazoles , Indoles/administración & dosificación , Masculino , Ratones , Ratones SCID , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico , Pirimidinas/administración & dosificación , Pirroles/administración & dosificación , Estudios Retrospectivos , Tumores Fibrosos Solitarios/irrigación sanguínea , Sulfonamidas/administración & dosificación , Sunitinib , Temozolomida , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Ann Oncol ; 24(7): 1931-1936, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23559153

RESUMEN

BACKGROUND: To report on a prospective, investigator-driven, phase II study on lapatinib in epidermal growth factor receptor (EGFR)-positive advanced chordoma patients. PATIENTS AND METHODS: From December 2009 to January 2012, 18 advanced progressing chordoma patients entered this study (median age: 61 years; disease extent: metastatic 72% and locally advanced 28%). Epidermal growth factor receptor (EGFR) expression and activation were evaluated by immunohistochemistry and/or phospho-arrays, real-time polimerase chain reaction, fluorescence immunostaining. Fluorescence in situ hybridization analysis was also carried out. Patients received lapatinib 1500 mg/day (mean dose intensity = 1282 mg/day), until progression or toxicity. The primary study end point was response rate (RR) as per Choi criteria. Secondary end points were RR by Response Evaluation Criteria in Solid Tumor (RECIST), overall survival, progression-free survival (PFS) and clinical benefit rate (CBR; RECIST complete response + partial response (PR) + stable disease (SD) ≥ 6 months). RESULTS: All patients were evaluable for response. Six (33.3%) patients had PR and 7 (38.9%) SD, as their best Choi responses, corresponding to RECIST SD in all cases. Median PFS by Choi was 6 [interquartile (IQ) range 3-8] months. Median PFS by RECIST was 8 (IQ range 4-12) months, with a 22% CBR. CONCLUSIONS: This phase II study showed a modest antitumor activity of lapatinib in chordoma. The clinical exploitation of EGFR targeting in chordoma needs to be further investigated, both clinically and preclinically. Clinical trial Registration No: EU Clinical Trials Register trial no. 2009-014456-29.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias Óseas/tratamiento farmacológico , Cordoma/tratamiento farmacológico , Receptores ErbB/metabolismo , Quinazolinas/administración & dosificación , Adulto , Anciano , Antineoplásicos/efectos adversos , Neoplasias Óseas/mortalidad , Neoplasias Óseas/patología , Cordoma/mortalidad , Cordoma/secundario , Supervivencia sin Enfermedad , Esquema de Medicación , Femenino , Humanos , Lapatinib , Masculino , Persona de Mediana Edad , Quinazolinas/efectos adversos , Sacro/patología , Base del Cráneo/patología , Resultado del Tratamiento
5.
Ann Oncol ; 22(7): 1682-1690, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21242589

RESUMEN

BACKGROUND: The purpose of this study was to confirm sunitinib activity in alveolar soft part sarcoma (ASPS) and to report on new insights into the molecular bases thereof. PATIENTS AND METHODS: From July 2007, nine patients with progressive metastatic ASPS received sunitinib 37.5 mg/day, within a named use program. Cryopreserved material was available for five naive patients, among whom three received sunitinib. Immunofluorescence (IF)/confocal microscopy, biochemical, and molecular/cytogenetic analyses were carried out, complemented by antiproliferative and activation assays in a short-term culture derived from one case. RESULTS: All patients were eligible for response. Best RECIST response was partial response in five cases, stable disease in three, and progression in one. The median progression-free survival was 17 months. Positron emission tomography results were consistent. Two cases of interval progressions were recorded. Antiproliferative assays and biochemistry on short-term culture showed that sunitinib is able to markedly impair ASPS cells growth and switch-off PDGFRB. IF/confocal microscopy demonstrated coexpression and physical association between PDGFRB/vascular endothelial growth factor receptor 2 (VEGFR2) and RET/VEGFR2 in ASPS cells, which was validated by biochemistry. PDGFRB, RET, and MET ligand-dependent activation was confirmed. CONCLUSIONS: We confirm the clinical efficacy of sunitinib in ASPS, mediated by PDGFRB, VEGFR2, and RET, which are all expressed in tumor cells. A direct antitumor effect was shown in a short-term cell culture.


Asunto(s)
Antineoplásicos/uso terapéutico , Proliferación Celular/efectos de los fármacos , Indoles/uso terapéutico , Pirroles/uso terapéutico , Sarcoma de Parte Blanda Alveolar/tratamiento farmacológico , Adulto , Western Blotting , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Estudios de Seguimiento , Humanos , Técnicas para Inmunoenzimas , Inmunoprecipitación , Hibridación Fluorescente in Situ , Masculino , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-ret/genética , Proteínas Proto-Oncogénicas c-ret/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Estudios Retrospectivos , Sarcoma de Parte Blanda Alveolar/genética , Sarcoma de Parte Blanda Alveolar/metabolismo , Sunitinib , Tasa de Supervivencia , Resultado del Tratamiento , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Adulto Joven
6.
J Pathol ; 217(1): 103-12, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18973210

RESUMEN

As the range of receptor tyrosine kinase (RTK) inhibitors widens, a detailed understanding of the activating mechanisms of KIT/platelet-derived growth factor receptor (PDGFR)A and the related downstream pathways involved in the development and maintenance of GISTs is becoming increasingly important. We analysed areas with different histological response ratios in surgical specimens taken from imatinib-treated and untreated GIST patients in order to investigate KIT and PDGFRA expression/activation, the presence of their cognate ligands and the activation of downstream signalling, by means of biochemistry, immunohistochemistry and flow cytometry. All of the cases showed KIT and PDGFRA co-expression. In addition to the oncogenic activation of mutated receptors, activation of wild-type KIT and wild-type PDGFRA, sustained by heterodimerization and an autocrine-paracrine loop, was demonstrated by the presence of their specific ligands, stem cell factor (SCF) and PDGFA. To confirm RTK activation further, all of the samples (including those with the highest regression ratios) were investigated for downstream effectors, and all proved to have activated downstream signalling. The results show that after the mutated receptors are switched off, heterologous wild-type receptors become important in imatinib-treated GISTs as a means of maintaining signalling activation. Taken together, our findings suggest that drugs targeting wild-type receptors should be tested in imatinib-treated GIST patients.


Asunto(s)
Antineoplásicos/uso terapéutico , Tumores del Estroma Gastrointestinal/metabolismo , Piperazinas/uso terapéutico , Proteínas Proto-Oncogénicas c-kit/metabolismo , Pirimidinas/uso terapéutico , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Benzamidas , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Tumores del Estroma Gastrointestinal/patología , Tumores del Estroma Gastrointestinal/cirugía , Humanos , Mesilato de Imatinib , Terapia Neoadyuvante , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Factor de Células Madre/metabolismo
7.
Br J Cancer ; 92(11): 1984-8, 2005 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-15900298

RESUMEN

The aim of the present study was to evaluate the effectiveness of two consecutive nonrandomised treatment programs applied between 1989 and 1999 at the Istituto Nazionale Tumori of Milan in an unselected cohort of 59 children over the age of one with stage 4 neuroblastoma. Both treatment programs consisted of two phases, the induction of the remission phase and the consolidation phase. The induction of the remission phase consisted of intensive chemotherapy, and remained the same throughout the study period. The consolidation phase consisted of sequential hemi-body irradiation (HBI) (10 Gy per session, 6 weeks apart) in the first period (1988-June 1994) and sequential high-dose cyclophosphamide, etoposide, mitoxantrone+L-PAM and autologous haemopoietic stem cell transplantation in the second (July 1994-1999). Intention-to-treat analysis revealed a significantly better outcome for patients treated with the second program, the 5-year event-free survival probability being 0.12 for program 1 and 0.31 for program 2 (P=0.03). This finding led us to conclude that sequential HBI is useless as consolidation treatment. The high-dose chemotherapy adopted in the second program enabled a proportion of patients to obtain long-term survival but, since the clinical results remain unsatisfactory, new treatment strategies are warranted.


Asunto(s)
Neuroblastoma/tratamiento farmacológico , Neuroblastoma/radioterapia , Trasplante de Células Madre , Adolescente , Niño , Preescolar , Terapia Combinada , Femenino , Humanos , Lactante , Masculino , Estadificación de Neoplasias , Neuroblastoma/patología , Análisis de Supervivencia , Trasplante Autólogo , Resultado del Tratamiento
8.
Blood ; 97(1): 81-8, 2001 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-11133745

RESUMEN

Severe combined immunodeficiency (SCID) comprises a heterogeneous group of primary immunodeficiencies, a proportion of which are due to mutations in either of the 2 recombination activating genes (RAG)-1 and -2, which mediate the process of V(D)J recombination leading to the assembly of antigen receptor genes. It is reported here that the clinical and immunologic phenotypes of patients bearing mutations in RAGs are more diverse than previously thought and that this variability is related, in part, to the specific type of RAG mutation. By analyzing 44 such patients from 41 families, the following conclusions were reached: (1) null mutations on both alleles lead to the T-B-SCID phenotype; (2) patients manifesting classic Omenn syndrome (OS) have missense mutations on at least one allele and maintain partial V(D)J recombination activity, which accounts for the generation of residual, oligoclonal T-lymphocytes; (3) in a third group of patients, findings were only partially compatible with OS, and these patients, who also carried at least one missense mutation, may be considered to have atypical SCID/OS; (4) patients with engraftment of maternal T cells as a complication of a transplacental transfusion represented a fourth group, and these patients, who often presented with a clinical phenotype mimicking OS, may be observed regardless of the type of RAG gene mutation. Analysis of the RAG genes by direct sequencing is an effective way to provide accurate diagnosis of RAG-deficient as opposed to RAG-independent V(D)J recombination defects, a distinction that cannot be made based on clinical and immunologic phenotype alone.


Asunto(s)
Genes RAG-1/genética , Región de Unión de la Inmunoglobulina/genética , Región Variable de Inmunoglobulina/genética , Linfocitos/inmunología , Alelos , Estudios de Cohortes , Análisis Mutacional de ADN , Proteínas de Unión al ADN/genética , Bases de Datos Factuales , Salud de la Familia , Femenino , Genotipo , Humanos , Inmunofenotipificación , Lactante , Recién Nacido , Linfopenia/etiología , Masculino , Intercambio Materno-Fetal/inmunología , Mutación , Mutación Missense , Proteínas Nucleares , Embarazo , Recombinación Genética , Inmunodeficiencia Combinada Grave/complicaciones , Inmunodeficiencia Combinada Grave/genética , Linfocitos T/trasplante
9.
Proc Natl Acad Sci U S A ; 97(26): 14572-7, 2000 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-11121059

RESUMEN

Omenn's syndrome is an autosomal recessive primary immunodeficiency characterized by variable numbers of T lymphocytes of limited clonality, hypereosinophilia, and high IgE levels with a paradoxical absence of circulating B lymphocytes. We have previously attributed this disorder to missense mutations that render the RAG1/RAG2 recombinase only partially active. Here we report seven Omenn's patients with a novel class of genetic lesions: frameshift mutations within the 5' coding region of RAG1. Interestingly, we demonstrate in transient expression experiments that these frameshift deletion alleles remain partially functional for both deletional and inversional recombination and can hence explain the partial rearrangement phenotype observed in these patients. The rearrangement activity is mediated by truncated RAG1 proteins that are generated by alternative ATG usage 3' to the frameshift deletion and that demonstrate improper cellular localization. Taken together, our results suggest a novel mechanism for the development of immunodeficiency in a subset of Omenn's syndrome patients.


Asunto(s)
ADN Nucleotidiltransferasas/fisiología , Mutación del Sistema de Lectura , Proteínas de Homeodominio/genética , Síndromes de Inmunodeficiencia/genética , Metionina/genética , Recombinación Genética , Alelos , Línea Celular Transformada , Proteínas de Unión al ADN/genética , Proteínas de Homeodominio/fisiología , Humanos , Metionina/fisiología , Proteínas Nucleares , Estructura Terciaria de Proteína , VDJ Recombinasas
10.
Mol Cell Biol ; 20(15): 5653-64, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10891502

RESUMEN

The V(D)J recombination reaction is composed of multiple nucleolytic processing steps mediated by the recombination-activating proteins RAG1 and RAG2. Sequence analysis has suggested that RAG2 contains six kelch repeat motifs that are predicted to form a six-bladed beta-propeller structure, with the second beta-strand of each repeat demonstrating marked conservation both within and between kelch repeat-containing proteins. Here we demonstrate that mutations G95R and DeltaI273 within the predicted second beta-strand of repeats 2 and 5 of RAG2 lead to immunodeficiency in patients P1 and P2. Green fluorescent protein fusions with the mutant proteins reveal appropriate localization to the nucleus. However, both mutations reduce the capacity of RAG2 to interact with RAG1 and block recombination signal cleavage, therefore implicating a defect in the early steps of the recombination reaction as the basis of the clinical phenotype. The present experiments, performed with an extensive panel of site-directed mutations within each of the six kelch motifs, further support the critical role of both hydrophobic and glycine-rich regions within the second beta-strand for RAG1-RAG2 interaction and recombination signal recognition and cleavage. In contrast, multiple mutations within the variable-loop regions of the kelch repeats had either mild or no effects on RAG1-RAG2 interaction and hence on the ability to mediate recombination. In all, the data demonstrate a critical role of the RAG2 kelch repeats for V(D)J recombination and highlight the importance of the conserved elements of the kelch motif.


Asunto(s)
Proteínas de Unión al ADN/genética , Síndromes de Inmunodeficiencia/genética , Mutación , Recombinación Genética , Secuencia de Aminoácidos , Línea Celular , Núcleo Celular/genética , Secuencia Conservada , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Humanos , Lactante , Masculino , Datos de Secuencia Molecular , Proteínas Nucleares , Conformación Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Secuencias Repetitivas de Aminoácido
11.
Prenat Diagn ; 20(1): 56-9, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10701853

RESUMEN

Mutations in recombination activating genes (RAG) 1 and 2 have been found to cause Omenn syndrome (OS), a severe combined immunodeficiency (SCID) with a peculiar phenotype. Here we report the prenatal diagnosis performed in three OS patients. Mutations were detected in the probands as well as in their parents by genomic sequencing of the complete coding regions of both RAG 1 and RAG 2, which are contained in a single exon. All the three probands had RAG 1 mutations in both alleles, at least one of which was a missense substitution. Of the three fetuses tested, one had a wild type sequence on both alleles, while the other two had one mutated allele. None of the three patients were predicted to be affected and this was confirmed at birth. Detection of RAG genes mutations on fetal samples by direct sequencing is an easy and effective way to investigate fetuses from families affected with RAG-dependent SCID and OS families affected by RAG-dependent SCID and OS.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Homeodominio/genética , Mutación , Diagnóstico Prenatal , Inmunodeficiencia Combinada Grave/diagnóstico , Amniocentesis , Secuencia de Bases , Muestra de la Vellosidad Coriónica , Análisis Mutacional de ADN , Femenino , Eliminación de Gen , Edad Gestacional , Humanos , Masculino , Mutación Missense , Proteínas Nucleares , Reacción en Cadena de la Polimerasa , Polimorfismo Conformacional Retorcido-Simple , Embarazo , Inmunodeficiencia Combinada Grave/genética , Síndrome
12.
Blood ; 94(10): 3468-78, 1999 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-10552957

RESUMEN

Mutations in the human RAG genes that impair, but do not abolish, recombination activity lead to Omenn syndrome, a severe primary immune deficiency that is associated with clinical and pathological features of graft-versus-host disease and oligoclonal expansion of activated, autologous T cells. We have analyzed the mechanisms accounting for peripheral oligoclonality of the T-cell repertoire. Predominance of few T-cell receptor clonotypes (both within TCRAB- and within TCRGD-expressing lymphocytes) is already detectable in the thymus and is further selected for in the periphery, with a different distribution of clonotypes in different tissues. These data indicate that oligoclonality of the T-cell repertoire in Omenn syndrome is due both to intrathymic restriction and to peripheral expansion. Moreover, the RAG genes defect that causes Omenn syndrome directly affects early stages of V(D)J recombination, but does not alter the process of double-strand-break DNA repair, including N and P nucleotide insertion.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Homeodominio/genética , Síndromes de Inmunodeficiencia/inmunología , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/inmunología , Secuencia de Bases , Femenino , Variación Genética , Humanos , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/patología , Recién Nacido , Leucocitos Mononucleares/inmunología , Datos de Secuencia Molecular , Mutación , Proteínas Nucleares , Homología de Secuencia de Ácido Nucleico , Timo/inmunología , Timo/patología
13.
J Clin Immunol ; 19(2): 87-97, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10226883

RESUMEN

In vertebrates, generation of the T- and B-cell repertoire relies on genomic rearrangement of T-cell receptor and immunoglobulin gene coding segments. This process, known as V(D)J recombination, is initiated by the lymphoid specific proteins Rag1 and Rag2. Both in humans and in animal models, mutations that abrogate expression of either the Rag1 or Rag2 proteins result in severe combined immune deficiency with a complete lack of circulating T and B cells due to an early block in lymphoid development. We have recently shown that mutations that impair, but do not completely abolish the function of Rag1 and Rag2 in humans result in Omenn syndrome, an enigmatic form of combined immune deficiency characterized by oligoclonal, activated T lymphocytes with a skewed Th2 profile.


Asunto(s)
Proteínas de Unión al ADN/genética , Genes RAG-1/genética , Recombinación Genética , Inmunodeficiencia Combinada Grave/genética , Animales , Secuencia de Bases , Proteínas de Unión al ADN/fisiología , Proteínas de Homeodominio/fisiología , Humanos , Datos de Secuencia Molecular , Mutación , Proteínas Nucleares , Inmunodeficiencia Combinada Grave/fisiopatología
14.
Mol Cell ; 4(6): 935-47, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10635319

RESUMEN

During V(D)J recombination, processing of branched coding end intermediates is essential for generating junctional diversity. Here, we report that the RAG1/ RAG2 recombinase is a 3' flap endonuclease. Substrates of this nuclease activity include various coding end intermediates, suggesting a direct role for RAG1/ RAG2 in generating junctional diversity during V(D)J recombination. Evidence is also provided indicating that site-specific RSS nicking involves RAG1/RAG2-mediated processing of a localized flap-like structure, implying 3' flap nicking in multiple DNA processing reactions. We have also demonstrated that the bacterial transposase Tn10 contains a 3' flap endonuclease activity, suggesting a mechanistic parallel between RAG1/RAG2 and other transposases. Based on these data, we propose that numerous transposases may facilitate genomic evolution by removing single-stranded extensions during the processing of excision site junctions.


Asunto(s)
Proteínas de Unión al ADN/genética , ADN/genética , Proteínas de Homeodominio/genética , Recombinación Genética , Animales , Secuencia de Bases , Variación Genética , Humanos , Ratones , Datos de Secuencia Molecular , Proteínas Nucleares , Transposasas/genética
15.
Br J Haematol ; 102(5): 1363-6, 1998 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9753072

RESUMEN

Severe combined immunodeficiency (SCID) comprises a heterogenous group of disorders that are fatal unless treated by bone marrow transplantation (BMT). The most common form of SCID (T-B+ SCID) is due to mutations of either the common gamma chain (gammac) or of gammac-coupled JAK3 kinase. We report an unusual JAK3 defect in a female who was successfully treated > 20 years ago with a BMT using her HLA-identical father as the donor. Persistence of genetically and biochemically defective autologous B cells, associated with reconstitution of cellular and humoral immunity, suggests that integrity of the gammac-JAK3 signalling pathway is not strictly required for immunoglobulin production.


Asunto(s)
Mutación , Proteínas Tirosina Quinasas/deficiencia , Inmunodeficiencia Combinada Grave/genética , Linfocitos B/patología , Western Blotting , Trasplante de Médula Ósea/métodos , Femenino , Eliminación de Gen , Humanos , Janus Quinasa 3 , Fosforilación , Proteínas Tirosina Quinasas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Inmunodeficiencia Combinada Grave/metabolismo , Inmunodeficiencia Combinada Grave/terapia , Linfocitos T/patología
16.
Cell ; 93(5): 885-96, 1998 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-9630231

RESUMEN

Genomic rearrangement of the antigen receptor loci is initiated by the two lymphoid-specific proteins Rag-1 and Rag-2. Null mutations in either of the two proteins abrogate initiation of V(D)J recombination and cause severe combined immunodeficiency with complete absence of mature B and T lymphocytes. We report here that patients with Omenn syndrome, a severe immunodeficiency characterized by the presence of activated, anergic, oligoclonal T cells, hypereosinophilia, and high IgE levels, bear missense mutations in either the Rag-1 or Rag-2 genes that result in partial activity of the two proteins. Two of the amino acid substitutions map within the Rag-1 homeodomain and decrease DNA binding activity, while three others lower the efficiency of Rag-1/Rag-2 interaction. These findings provide evidence to indicate that the immunodeficiency manifested in patients with Omenn syndrome arises from mutations that decrease the efficiency of V(D)J recombination.


Asunto(s)
Reordenamiento Génico de Linfocito T/genética , Proteínas de Homeodominio/genética , Mutación , Receptores de Antígenos de Linfocitos T/genética , Recombinación Genética , Inmunodeficiencia Combinada Grave/etiología , Secuencia de Aminoácidos , Anergia Clonal , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Eosinofilia , Femenino , Genes RAG-1 , Genotipo , Proteínas de Homeodominio/metabolismo , Humanos , Activación de Linfocitos , Datos de Secuencia Molecular , Proteínas Nucleares , Unión Proteica , Inmunodeficiencia Combinada Grave/genética , Síndrome
17.
Blood ; 90(10): 3996-4003, 1997 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-9354668

RESUMEN

Mutations of the Janus family kinase JAK3 have been found to be responsible for autosomal recessive severe combined immunodeficiency (SCID) in humans. We report here the analysis of four new unrelated patients affected by JAK3-deficient SCID. The genetic defects were heterogeneous and included a large intragenic deletion as well as different point mutations, leading to missense substitutions, early stop codons, or splicing defects. We performed a series of studies of the biochemical events induced by cytokines on lymphoblastoid B-cell lines obtained from these patients. Abnormalities in tyrosine phosphorylation of JAK3 in response to interleukin-2 (IL-2) and IL-4 were present in all patients. Accordingly, IL-2-mediated phosphorylation of STAT5 was also absent or barely detectable. On the contrary, in all cases, we could show reduced but clear phosphorylation of STAT6 upon IL-4 stimulation. In one patient carrying a single amino acid change (Glu481Gly) in the JH3 domain of JAK3, we observed partially conserved IL-2 responses resulting in reduced but detectable levels of JAK3 and STAT5 phosphorylation. Interestingly, the patient bearing this mutation developed a substantial number of circulating CD4(+)/CD45RO+ activated T lymphocytes that were functionally impaired. In two cases, patients' cells expressed JAK3 proteins with mutations in the JH2 pseudo-kinase domain. A single cysteine to arginine substitution (Cys759Arg) in this region resulted in high basal levels of constitutive JAK3 tyrosine phosphorylation unresponsive to either downregulation by serum starvation or cytokine-mediated upregulation. The characterization of the genetic defects and biochemical abnormalities in these JAK3-deficient patients will help define the role of JAK3 in the ontogeny of a competent immune system and may lead to a better understanding of the JAK3 functional domains.


Asunto(s)
Linfocitos B/metabolismo , Mutación , Proteínas Tirosina Quinasas/genética , Inmunodeficiencia Combinada Grave/genética , Células Cultivadas , Humanos , Interleucina-2/genética , Interleucina-2/metabolismo , Interleucina-4/genética , Interleucina-4/metabolismo , Janus Quinasa 3 , Proteínas Tirosina Quinasas/deficiencia , Inmunodeficiencia Combinada Grave/enzimología
18.
Biochem Biophys Res Commun ; 229(2): 494-7, 1996 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-8954926

RESUMEN

We have determined the genomic structure of the human LHX1 gene, a member of the LIM/homeobox (Lhx) gene family. The transcript is assembled from five exons, which are separated by introns ranging in size from 93 nt to 2.3 kb. The two LIM domains are entirely contained in the first and second exons, respectively, while the homeodomain is split into exons three and four. This structure closely parallels the organization of other mouse and human Lhx genes whose genomic structure is known. An exception is the mouse and human is/1 genes, whose homeodomain does not contain introns. An intron at the same position also occurs in the Xlim 1 gene as well as in other homeobox genes, such as evx1 and evx2, suggesting that this intron insertion represents an ancestral event, from which homeobox genes of different families originated. In this context, evolution of the Lhx gene family probably involved the shuffling of this intron-containing homeobox in the proximity of a LIM-only gene, while Islet genes were formed either by the shuffling of an intronless homeobox to the same LIM domain or, alternatively, by intron loss during their evolution.


Asunto(s)
Exones , Proteínas de Homeodominio/genética , Intrones , Animales , Clonación Molecular , Genes Homeobox , Humanos , Proteínas con Homeodominio LIM , Ratones , Datos de Secuencia Molecular , Factores de Transcripción
19.
Genomics ; 35(2): 312-20, 1996 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-8661144

RESUMEN

We have previously reported (Villa et al. (1993), Genomics 18: 223) the characterization of the human ZNF75 gene located on Xq26, which has only limited homology (less than 65%) to other ZF genes in the databases. Here, we describe three human zinc finger genes with 86 to 95% homology to ZNF75 at the nucleotide level, which represent all the members of the human ZNF75 subfamily. One of these, ZNF75B, is a pseudogene mapped to chromosome 12q13. The other two, ZNF75A and ZNF75C, maintain an ORF in the sequenced region, and at least the latter is expressed in the U937 cell line. They were mapped to chromosomes 16 and 11, respectively. All these genes are conserved in chimpanzees, gorillas, and orangutans. The ZNF75B homologue is a pseudogene in all three great apes, and in chimpanzee it is located on chromosome 10 (phylogenetic XII), at p13 (corresponding to the human 12q13). The chimpanzee homologue of ZNF75 is also located on the Xq26 chromosome, in the same region, as detected by in situ hybridization. As expected, nucleotide changes were clearly more abundant between human and orangutan than between human and chimpanzee or gorilla homologues. Members of the same class were more similar to each other than to the other homologues within the same species. This suggests that the duplication and/or retrotranscription events occurred in a common ancestor long before great ape speciation. This, together with the existence of at least two genes in cows and horses, suggests a relatively high conservation of this gene family.


Asunto(s)
Cromosomas Humanos Par 12 , Cromosomas Humanos Par 16 , Cromosomas Humanos Par 17 , Proteínas de Unión al ADN/genética , Familia de Multigenes , Pongo pygmaeus/genética , Factores de Transcripción/genética , Dedos de Zinc/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Bovinos , Mapeo Cromosómico , Secuencia Conservada , Cartilla de ADN , Proteínas de Unión al ADN/biosíntesis , Gorilla gorilla/genética , Caballos , Humanos , Factores de Transcripción de Tipo Kruppel , Masculino , Mamíferos , Ratones , Datos de Secuencia Molecular , Pan troglodytes/genética , Reacción en Cadena de la Polimerasa , Seudogenes , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico , Factores de Transcripción/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA