Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 100(5): 725-736, 2017 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-28475857

RESUMEN

To explore the genetic architecture of human overgrowth syndromes and human growth control, we performed experimental and bioinformatic analyses of 710 individuals with overgrowth (height and/or head circumference ≥+2 SD) and intellectual disability (OGID). We identified a causal mutation in 1 of 14 genes in 50% (353/710). This includes HIST1H1E, encoding histone H1.4, which has not been associated with a developmental disorder previously. The pathogenic HIST1H1E mutations are predicted to result in a product that is less effective in neutralizing negatively charged linker DNA because it has a reduced net charge, and in DNA binding and protein-protein interactions because key residues are truncated. Functional network analyses demonstrated that epigenetic regulation is a prominent biological process dysregulated in individuals with OGID. Mutations in six epigenetic regulation genes-NSD1, EZH2, DNMT3A, CHD8, HIST1H1E, and EED-accounted for 44% of individuals (311/710). There was significant overlap between the 14 genes involved in OGID and 611 genes in regions identified in GWASs to be associated with height (p = 6.84 × 10-8), suggesting that a common variation impacting function of genes involved in OGID influences height at a population level. Increased cellular growth is a hallmark of cancer and there was striking overlap between the genes involved in OGID and 260 somatically mutated cancer driver genes (p = 1.75 × 10-14). However, the mutation spectra of genes involved in OGID and cancer differ, suggesting complex genotype-phenotype relationships. These data reveal insights into the genetic control of human growth and demonstrate that exome sequencing in OGID has a high diagnostic yield.


Asunto(s)
Discapacidades del Desarrollo/genética , Epigénesis Genética , Discapacidad Intelectual/genética , Mutación , Adolescente , Secuencia de Aminoácidos , Niño , Preescolar , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Proteínas de Unión al ADN/genética , Discapacidades del Desarrollo/diagnóstico , Proteína Potenciadora del Homólogo Zeste 2/genética , Femenino , Regulación de la Expresión Génica , Sitios Genéticos , Estudio de Asociación del Genoma Completo , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina , Histonas/genética , Humanos , Lactante , Discapacidad Intelectual/diagnóstico , Péptidos y Proteínas de Señalización Intracelular/genética , Desequilibrio de Ligamiento , Masculino , Neoplasias/diagnóstico , Neoplasias/genética , Proteínas Nucleares/genética , Análisis de Secuencia de ADN , Factores de Transcripción/genética
2.
PLoS Genet ; 13(5): e1006787, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28545128

RESUMEN

Neuroblastoma is a cancer of the developing sympathetic nervous system that most commonly presents in young children and accounts for approximately 12% of pediatric oncology deaths. Here, we report on a genome-wide association study (GWAS) in a discovery cohort or 2,101 cases and 4,202 controls of European ancestry. We identify two new association signals at 3q25 and 4p16 that replicated robustly in multiple independent cohorts comprising 1,163 cases and 4,396 controls (3q25: rs6441201 combined P = 1.2x10-11, Odds Ratio 1.23, 95% CI:1.16-1.31; 4p16: rs3796727 combined P = 1.26x10-12, Odds Ratio 1.30, 95% CI: 1.21-1.40). The 4p16 signal maps within the carboxypeptidase Z (CPZ) gene. The 3q25 signal resides within the arginine/serine-rich coiled-coil 1 (RSRC1) gene and upstream of the myeloid leukemia factor 1 (MLF1) gene. Increased expression of MLF1 was observed in neuroblastoma cells homozygous for the rs6441201 risk allele (P = 0.02), and significant growth inhibition was observed upon depletion of MLF1 (P < 0.0001) in neuroblastoma cells. Taken together, we show that common DNA variants within CPZ at 4p16 and upstream of MLF1 at 3q25 influence neuroblastoma susceptibility and MLF1 likely plays an important role in neuroblastoma tumorigenesis.


Asunto(s)
Carboxipeptidasas/genética , Cromosomas Humanos Par 3/genética , Cromosomas Humanos Par 4/genética , Neuroblastoma/genética , Polimorfismo de Nucleótido Simple , Proteínas/genética , Estudios de Casos y Controles , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Proteínas de Unión al ADN , Femenino , Silenciador del Gen , Homocigoto , Humanos , Masculino , Proteínas Nucleares/genética , Proteínas/metabolismo
3.
Am J Med Genet C Semin Med Genet ; 181(4): 638-643, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31714006

RESUMEN

BRWD3 has been described as a cause of X-linked intellectual disability, but relatively little is known about the specific phenotype. We report the largest BRWD3 patient series to date, comprising 17 males with 12 distinct null variants and 2 partial gene deletions. All patients presented with intellectual disability, which was classified as moderate (65%) or mild (35%). Behavioral issues were present in 75% of patients, including aggressive behavior, attention deficit/hyperactivity and/or autistic spectrum disorders. Mean head circumference was +2.8 SD (2.8 standard deviations above the mean), and mean BMI was +2.0 SD (in the context of a mean height of +1.3 SD), indicating a predominant macrocephaly/obesity phenotype. Shared facial features included a tall chin, prognathism, broad forehead, and prominent supraorbital ridge. Additional features, reported in a minority (<30%) of patients included cryptorchidism, neonatal hypotonia, and small joint hypermobility. This study delineates the clinical features associated with BRWD3 null variants and partial gene deletions, and suggests that BRWD3 should be included in the differential diagnosis of patients with an overgrowth-intellectual disability (OGID) phenotype, particularly in male patients with a mild or moderate intellectual disability associated with macrocephaly and/or obesity.


Asunto(s)
Eliminación de Gen , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Discapacidad Intelectual/genética , Megalencefalia/genética , Obesidad/genética , Factores de Transcripción/genética , Adolescente , Niño , Humanos , Masculino , Índice de Severidad de la Enfermedad , Síndrome
4.
Am J Med Genet C Semin Med Genet ; 181(4): 502-508, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31479583

RESUMEN

Sotos syndrome is an overgrowth-intellectual disability (OGID) syndrome caused by NSD1 pathogenic variants and characterized by a distinctive facial appearance, an intellectual disability, tall stature and/or macrocephaly. Other associated clinical features include scoliosis, seizures, renal anomalies, and cardiac anomalies. However, many of the published Sotos syndrome clinical descriptions are based on studies of children; the phenotype in adults with Sotos syndrome is not yet well described. Given that it is now 17 years since disruption of NSD1 was shown to cause Sotos syndrome, many of the children first reported are now adults. It is therefore timely to investigate the phenotype of 44 adults with Sotos syndrome and NSD1 pathogenic variants. We have shown that adults with Sotos syndrome display a wide spectrum of intellectual ability with functioning ranging from fully independent to fully dependent. Reproductive rates are low. In our cohort, median height in adult women is +1.9 SD and men +0.5 SD. There is a distinctive facial appearance in adults with a tall, square, prominent chin. Reassuringly, adults with Sotos syndrome are generally healthy with few new medical issues; however, lymphedema, poor dentition, hearing loss, contractures and tremor have developed in a small number of individuals.


Asunto(s)
Fenotipo , Síndrome de Sotos/fisiopatología , Adulto , Niño , Facies , Femenino , Humanos , Discapacidad Intelectual/genética , Masculino , Síndrome de Sotos/genética , Síndrome de Sotos/psicología
5.
Am J Med Genet C Semin Med Genet ; 181(4): 557-564, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31721432

RESUMEN

CHD8 has been reported as an autism susceptibility/intellectual disability gene but emerging evidence suggests that it additionally causes an overgrowth phenotype. This study reports 27 unrelated patients with pathogenic or likely pathogenic CHD8 variants (25 null variants, two missense variants) and a male:female ratio of 21:6 (3.5:1, p < .01). All patients presented with intellectual disability, with 85% in the mild or moderate range, and 85% had a height and/or head circumference ≥2 standard deviations above the mean, meeting our clinical criteria for overgrowth. Behavioral problems were reported in the majority of patients (78%), with over half (56%) either formally diagnosed with an autistic spectrum disorder or described as having autistic traits. Additional clinical features included neonatal hypotonia (33%), and less frequently seizures, pes planus, scoliosis, fifth finger clinodactyly, umbilical hernia, and glabellar hemangioma (≤15% each). These results suggest that, in addition to its established link with autism and intellectual disability, CHD8 causes an overgrowth phenotype, and should be considered in the differential diagnosis of patients presenting with increased height and/or head circumference in association with intellectual disability.


Asunto(s)
Cadherinas/genética , Trastornos del Crecimiento/genética , Fenotipo , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Lactante , Discapacidad Intelectual/genética , Masculino , Síndrome , Adulto Joven
6.
Am J Med Genet A ; 179(4): 588-594, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30793471

RESUMEN

Overgrowth-intellectual disability (OGID) syndromes are characterized by increased growth (height and/or head circumference ≥+2 SD) in association with an intellectual disability. Constitutive EED variants have previously been reported in five individuals with an OGID syndrome, eponymously designated Cohen-Gibson syndrome and resembling Weaver syndrome. Here, we report three additional individuals with constitutive EED variants, identified through exome sequencing of an OGID patient series. We compare the EED phenotype with that of Weaver syndrome (56 individuals), caused by constitutive EZH2 variants. We conclude that while there is considerable overlap between the EED and EZH2 phenotypes with both characteristically associated with increased growth and an intellectual disability, individuals with EED variants more frequently have cardiac problems and cervical spine abnormalities, boys have cryptorchidism and the facial gestalts can usually be distinguished.


Asunto(s)
Anomalías Múltiples/patología , Hipotiroidismo Congénito/patología , Anomalías Craneofaciales/patología , Proteína Potenciadora del Homólogo Zeste 2/genética , Dedos/anomalías , Trastornos del Crecimiento/patología , Deformidades Congénitas de la Mano/patología , Discapacidad Intelectual/patología , Microcefalia/patología , Hipotonía Muscular/patología , Mutación , Miopía/patología , Obesidad/patología , Complejo Represivo Polycomb 2/genética , Degeneración Retiniana/patología , Anomalías Múltiples/genética , Adulto , Niño , Hipotiroidismo Congénito/genética , Anomalías Craneofaciales/genética , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Femenino , Dedos/patología , Trastornos del Crecimiento/genética , Deformidades Congénitas de la Mano/genética , Humanos , Discapacidad Intelectual/genética , Masculino , Microcefalia/genética , Hipotonía Muscular/genética , Miopía/genética , Obesidad/genética , Fenotipo , Degeneración Retiniana/genética , Secuenciación del Exoma , Adulto Joven
7.
Am J Med Genet A ; 179(10): 2049-2055, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31400068

RESUMEN

Histone Gene Cluster 1 Member E, HIST1H1E, encodes Histone H1.4, is one of a family of epigenetic regulator genes, acts as a linker histone protein, and is responsible for higher order chromatin structure. HIST1H1E syndrome (also known as Rahman syndrome, OMIM #617537) is a recently described intellectual disability (ID) syndrome. Since the initial description of five unrelated individuals with three different heterozygous protein-truncating variants (PTVs) in the HIST1H1E gene in 2017, we have recruited 30 patients, all with HIST1H1E PTVs that result in the same shift in frame and that cluster to a 94-base pair region in the HIST1H1E carboxy terminal domain. The identification of 30 patients with HIST1H1E variants has allowed the clarification of the HIST1H1E syndrome phenotype. Major findings include an ID and a recognizable facial appearance. ID was reported in all patients and is most frequently of moderate severity. The facial gestalt consists of a high frontal hairline and full lower cheeks in early childhood and, in later childhood and adulthood, affected individuals have a strikingly high frontal hairline, frontal bossing, and deep-set eyes. Other associated clinical features include hypothyroidism, abnormal dentition, behavioral issues, cryptorchidism, skeletal anomalies, and cardiac anomalies. Brain magnetic resonance imaging (MRI) is frequently abnormal with a slender corpus callosum a frequent finding.


Asunto(s)
Facies , Histonas/genética , Discapacidad Intelectual/genética , Mutación/genética , Conducta , Crecimiento y Desarrollo , Heterocigoto , Humanos , Aprendizaje , Fenotipo , Síndrome
8.
Nature ; 493(7432): 406-10, 2013 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-23242139

RESUMEN

Improved sequencing technologies offer unprecedented opportunities for investigating the role of rare genetic variation in common disease. However, there are considerable challenges with respect to study design, data analysis and replication. Using pooled next-generation sequencing of 507 genes implicated in the repair of DNA in 1,150 samples, an analytical strategy focused on protein-truncating variants (PTVs) and a large-scale sequencing case-control replication experiment in 13,642 individuals, here we show that rare PTVs in the p53-inducible protein phosphatase PPM1D are associated with predisposition to breast cancer and ovarian cancer. PPM1D PTV mutations were present in 25 out of 7,781 cases versus 1 out of 5,861 controls (P = 1.12 × 10(-5)), including 18 mutations in 6,912 individuals with breast cancer (P = 2.42 × 10(-4)) and 12 mutations in 1,121 individuals with ovarian cancer (P = 3.10 × 10(-9)). Notably, all of the identified PPM1D PTVs were mosaic in lymphocyte DNA and clustered within a 370-base-pair region in the final exon of the gene, carboxy-terminal to the phosphatase catalytic domain. Functional studies demonstrate that the mutations result in enhanced suppression of p53 in response to ionizing radiation exposure, suggesting that the mutant alleles encode hyperactive PPM1D isoforms. Thus, although the mutations cause premature protein truncation, they do not result in the simple loss-of-function effect typically associated with this class of variant, but instead probably have a gain-of-function effect. Our results have implications for the detection and management of breast and ovarian cancer risk. More generally, these data provide new insights into the role of rare and of mosaic genetic variants in common conditions, and the use of sequencing in their identification.


Asunto(s)
Neoplasias de la Mama/genética , Predisposición Genética a la Enfermedad/genética , Mosaicismo , Mutación , Neoplasias Ováricas/genética , Fosfoproteínas Fosfatasas/genética , Alelos , Análisis por Conglomerados , Exones , Femenino , Humanos , Isoenzimas/genética , Linfocitos/metabolismo , Proteína Fosfatasa 2C , Análisis de Secuencia de ADN , Proteína p53 Supresora de Tumor/metabolismo
9.
Mol Cell ; 43(3): 432-48, 2011 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-21737329

RESUMEN

A better understanding of the mechanisms through which anticancer drugs exert their effects is essential to improve combination therapies. While studying how genotoxic stress kills cancer cells, we discovered a large ∼2MDa cell death-inducing platform, referred to as "Ripoptosome." It contains the core components RIP1, FADD, and caspase-8, and assembles in response to genotoxic stress-induced depletion of XIAP, cIAP1 and cIAP2. Importantly, it forms independently of TNF, CD95L/FASL, TRAIL, death-receptors, and mitochondrial pathways. It also forms upon Smac-mimetic (SM) treatment without involvement of autocrine TNF. Ripoptosome assembly requires RIP1's kinase activity and can stimulate caspase-8-mediated apoptosis as well as caspase-independent necrosis. It is negatively regulated by FLIP, cIAP1, cIAP2, and XIAP. Mechanistically, IAPs target components of this complex for ubiquitylation and inactivation. Moreover, we find that etoposide-stimulated Ripoptosome formation converts proinflammatory cytokines into prodeath signals. Together, our observations shed new light on fundamental mechanisms by which chemotherapeutics may kill cancer cells.


Asunto(s)
Apoptosis/fisiología , Caspasa 8/fisiología , Daño del ADN , Proteína de Dominio de Muerte Asociada a Fas/fisiología , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas de Complejo Poro Nuclear/fisiología , Proteínas de Unión al ARN/fisiología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/fisiología , Caspasa 8/química , Caspasa 8/metabolismo , Línea Celular Tumoral , Activación Enzimática , Etopósido/farmacología , Proteína de Dominio de Muerte Asociada a Fas/química , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Humanos , Proteínas Inhibidoras de la Apoptosis/fisiología , Ligandos , Mitocondrias/metabolismo , Proteínas de Complejo Poro Nuclear/química , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , Transducción de Señal
10.
Hum Mol Genet ; 24(17): 4775-9, 2015 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-25972378

RESUMEN

Overgrowth syndromes comprise a group of heterogeneous disorders characterised by excessive growth parameters, often in association with intellectual disability. To identify new causes of human overgrowth, we have been undertaking trio-based exome sequencing studies in overgrowth patients and their unaffected parents. Prioritisation of functionally relevant genes with multiple unique de novo mutations revealed four mutations in protein phosphatase 2A (PP2A) regulatory subunit B family genes protein phosphatase 2, regulatory Subunit B', beta (PPP2R5B); protein phosphatase 2, regulatory Subunit B', gamma (PPP2R5C); and protein phosphatase 2, regulatory Subunit B', delta (PPP2R5D). This observation in 3 related genes in 111 individuals with a similar phenotype is greatly in excess of the expected number, as determined from gene-specific de novo mutation rates (P = 1.43 × 10(-10)). Analysis of exome-sequencing data from a follow-up series of overgrowth probands identified a further pathogenic mutation, bringing the total number of affected individuals to 5. Heterozygotes shared similar phenotypic features including increased height, increased head circumference and intellectual disability. The mutations clustered within a region of nine amino acid residues in the aligned protein sequences (P = 1.6 × 10(-5)). We mapped the mutations onto the crystal structure of the PP2A holoenzyme complex to predict their molecular and functional consequences. These studies suggest that the mutations may affect substrate binding, thus perturbing the ability of PP2A to dephosphorylate particular protein substrates. PP2A is a major negative regulator of v-akt murine thymoma viral oncogene homolog 1 (AKT). Thus, our data further expand the list of genes encoding components of the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/AKT signalling cascade that are disrupted in human overgrowth conditions.


Asunto(s)
Estudios de Asociación Genética , Trastornos del Crecimiento/genética , Proteínas de la Membrana/genética , Mutación , Proteína Fosfatasa 2/genética , Exoma , Humanos , Proteínas de la Membrana/química , Modelos Moleculares , Fenotipo , Conformación Proteica , Proteína Fosfatasa 2/química
12.
Nat Cell Biol ; 7(1): 70-7, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15580265

RESUMEN

Some members of the inhibitor of apoptosis (IAP) family suppress apoptosis by neutralizing caspases. The current model suggests that all caspase-regulatory IAPs function as direct enzyme inhibitors, blocking effector caspases by binding to their catalytically active pockets. Here we show that IAPs are functionally non-equivalent and regulate effector caspases through distinct mechanisms. Whereas XIAP binds directly to the active-site pockets of effector caspases, we find that regulation of effector caspases by Drosophila IAP1 (DIAP1) requires an evolutionarily conserved IAP-binding motif (IBM) at the neo-amino terminus of the large caspase subunit. Remarkably, unlike XIAP, DIAP1-sequestered effector caspases remain catalytically active, suggesting that DIAP1 does not function as a bona fide enzyme inhibitor. Moreover, we demonstrate that the mammalian IAP c-IAP1 interacts with caspase-7 in an exclusively IBM-dependent, but active site pocket-independent, manner that is mechanistically similar to DIAP1. The importance of IBM-mediated regulation of effector-caspases in vivo is substantiated by the enhanced apoptotic potency of IBM-mutant versions of drICE, DCP-1 and caspase-7.


Asunto(s)
Apoptosis/fisiología , Caspasas/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas/metabolismo , Transducción de Señal/fisiología , Secuencias de Aminoácidos/fisiología , Animales , Sitios de Unión/fisiología , Caspasa 7 , Caspasas/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Humanos , Proteínas Inhibidoras de la Apoptosis , Ratones , Mutación/genética , Células 3T3 NIH , Unión Proteica/fisiología , Subunidades de Proteína/metabolismo , Ubiquitina-Proteína Ligasas , Proteína Inhibidora de la Apoptosis Ligada a X
13.
Clin Cancer Res ; 28(21): 4634-4641, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-35984704

RESUMEN

PURPOSE: CT900 is a novel small molecule thymidylate synthase inhibitor that binds to α-folate receptor (α-FR) and thus is selectively taken up by α-FR-overexpressing tumors. PATIENTS AND METHODS: A 3+3 dose escalation design was used. During dose escalation, CT900 doses of 1-6 mg/m2 weekly and 2-12 mg/m2 every 2 weeks (q2Wk) intravenously were evaluated. Patients with high-grade serous ovarian cancer were enrolled in the expansion cohorts. RESULTS: 109 patients were enrolled: 42 patients in the dose escalation and 67 patients in the expansion cohorts. At the dose/schedule of 12 mg/m2/q2Wk (with and without dexamethasone, n = 40), the most common treatment-related adverse events were fatigue, nausea, diarrhea, cough, anemia, and pneumonitis, which were predominantly grade 1 and grade 2. Levels of CT900 more than 600 nmol/L needed for growth inhibition in preclinical models were achieved for >65 hours at a dose of 12 mg/m2. In the expansion cohorts, the overall response rate (ORR), was 14/64 (21.9%). Thirty-eight response-evaluable patients in the expansion cohorts receiving 12 mg/m2/q2Wk had tumor evaluable for quantification of α-FR. Patients with high or medium expression had an objective response rate of 9/25 (36%) compared with 1/13 (7.7%) in patients with negative/very low or low expression of α-FR. CONCLUSIONS: The dose of 12 mg/m2/q2Wk was declared the recommended phase II dose/schedule. At this dose/schedule, CT900 exhibited an acceptable side effect profile with clinical benefit in patients with high/medium α-FR expression and warrants further investigation.


Asunto(s)
Neoplasias , Neoplasias Ováricas , Humanos , Femenino , Timidilato Sintasa/genética , Dosis Máxima Tolerada , Neoplasias/tratamiento farmacológico , Inhibidores Enzimáticos/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Ácido Fólico
14.
J Immunother Cancer ; 10(6)2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35717027

RESUMEN

BACKGROUND: Data suggest that immunomodulation induced by DNA hypomethylating agents can sensitize tumors to immune checkpoint inhibitors. We conducted a phase 1 dose-escalation trial (NCT02998567) of guadecitabine and pembrolizumab in patients with advanced solid tumors. We hypothesized that guadecitabine will overcome pembrolizumab resistance. METHODS: Patients received guadecitabine (45 mg/m2 or 30 mg/m2, administered subcutaneously on days 1-4), with pembrolizumab (200 mg administered intravenously starting from cycle 2 onwards) every 3 weeks. Primary endpoints were safety, tolerability and maximum tolerated dose; secondary and exploratory endpoints included objective response rate (ORR), changes in methylome, transcriptome, immune contextures in pre-treatment and on-treatment tumor biopsies. RESULTS: Between January 2017 and January 2020, 34 patients were enrolled. The recommended phase II dose was guadecitabine 30 mg/m2, days 1-4, and pembrolizumab 200 mg on day 1 every 3 weeks. Two dose-limiting toxicities (neutropenia, febrile neutropenia) were reported at guadecitabine 45 mg/m2 with none reported at guadecitabine 30 mg/m2. The most common treatment-related adverse events (TRAEs) were neutropenia (58.8%), fatigue (17.6%), febrile neutropenia (11.8%) and nausea (11.8%). Common, grade 3+ TRAEs were neutropaenia (38.2%) and febrile neutropaenia (11.8%). There were no treatment-related deaths. Overall, 30 patients were evaluable for antitumor activity; ORR was 7% with 37% achieving disease control (progression-free survival) for ≥24 weeks. Of 12 evaluable patients with non-small cell lung cancer, 10 had been previously treated with immune checkpoint inhibitors with 5 (42%) having disease control ≥24 weeks (clinical benefit). Reduction in LINE-1 DNA methylation following treatment in blood (peripheral blood mononuclear cells) and tissue samples was demonstrated and methylation at transcriptional start site and 5' untranslated region gene regions showed enriched negative correlation with gene expression. Increases in intra-tumoural effector T-cells were seen in some responding patients. Patients having clinical benefit had high baseline inflammatory signature on RNAseq analyses. CONCLUSIONS: Guadecitabine in combination with pembrolizumab is tolerable with biological and anticancer activity. Reversal of previous resistance to immune checkpoint inhibitors is demonstrated.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Neoplasias , Anticuerpos Monoclonales Humanizados/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Azacitidina/análogos & derivados , Azacitidina/uso terapéutico , Humanos , Inhibidores de Puntos de Control Inmunológico , Neoplasias/tratamiento farmacológico
15.
Nat Cell Biol ; 5(5): 467-73, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12692559

RESUMEN

Some members of the inhibitor of apoptosis (IAP) protein family block apoptosis by binding to and neutralizing active caspases. We recently demonstrated that a physical association between IAP and caspases alone is insufficient to regulate caspases in vivo and that an additional level of control is provided by IAP-mediated ubiquitination of both itself and the associated caspases. Here we show that Drosophila IAP 1 (DIAP1) is degraded by the 'N-end rule' pathway and that this process is indispensable for regulating apoptosis. Caspase-mediated cleavage of DIAP1 at position 20 converts the more stable pro-N-degron of DIAP1 into the highly unstable, Asn-bearing, DIAP1 N-degron of the N-end rule degradation pathway. Thus, DIAP1 represents the first known metazoan substrate of the N-end rule pathway that is targeted for degradation through its amino-terminal Asn residue. We demonstrate that the N-end rule pathway is required for regulation of apoptosis induced by Reaper and Hid expression in the Drosophila melanogaster eye. Our data suggest that DIAP1 instability, mediated through caspase activity and subsequent exposure of the N-end rule pathway, is essential for suppression of apoptosis. We suggest that DIAP1 safeguards cell viability through the coordinated mutual destruction of itself and associated active caspases.


Asunto(s)
Apoptosis/genética , Proteínas de Drosophila/deficiencia , Drosophila melanogaster/metabolismo , Anomalías del Ojo/metabolismo , Ojo/metabolismo , Transducción de Señal/genética , Secuencia de Aminoácidos/fisiología , Animales , Asparagina/metabolismo , Caspasas/metabolismo , Supervivencia Celular/fisiología , Células Cultivadas , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Ojo/citología , Anomalías del Ojo/genética , Eliminación de Gen , Proteínas Inhibidoras de la Apoptosis , Mutación/genética , Neuropéptidos/metabolismo , Fenotipo , Estructura Terciaria de Proteína/fisiología
16.
Nat Cell Biol ; 4(6): 445-50, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12021771

RESUMEN

Members of the Inhibitor of Apoptosis Protein (IAP) family block activation of the intrinsic cell death machinery by binding to and neutralizing the activity of pro-apoptotic caspases. In Drosophila melanogaster, the pro-apoptotic proteins Reaper (Rpr), Grim and Hid (head involution defective) all induce cell death by antagonizing the anti-apoptotic activity of Drosophila IAP1 (DIAP1), thereby liberating caspases. Here, we show that in vivo, the RING finger of DIAP1 is essential for the regulation of apoptosis induced by Rpr, Hid and Dronc. Furthermore, we show that the RING finger of DIAP1 promotes the ubiquitination of both itself and of Dronc. Disruption of the DIAP1 RING finger does not inhibit its binding to Rpr, Hid or Dronc, but completely abrogates ubiquitination of Dronc. Our data suggest that IAPs suppress apoptosis by binding to and targeting caspases for ubiquitination.


Asunto(s)
Apoptosis/fisiología , Caspasas/metabolismo , Proteínas de Drosophila/metabolismo , Ubiquitina/metabolismo , Animales , Células Cultivadas , Drosophila , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Proteínas Inhibidoras de la Apoptosis , Ligasas/metabolismo , Mutagénesis/fisiología , Neuropéptidos/metabolismo , Péptidos/metabolismo , Células Fotorreceptoras de Invertebrados/citología , Células Fotorreceptoras de Invertebrados/enzimología , Unión Proteica/fisiología , Estructura Terciaria de Proteína , Retina/citología , Retina/enzimología , Ubiquitina-Proteína Ligasas
17.
Lancet Child Adolesc Health ; 3(5): 322-331, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30885698

RESUMEN

BACKGROUND: Wilms tumour is the most common childhood renal cancer and is genetically heterogeneous. While several Wilms tumour predisposition genes have been identified, there is strong evidence that further predisposition genes are likely to exist. Our study aim was to identify new predisposition genes for Wilms tumour. METHODS: In this exome sequencing study, we analysed lymphocyte DNA from 890 individuals with Wilms tumour, including 91 affected individuals from 49 familial Wilms tumour pedigrees. We used the protein-truncating variant prioritisation method to prioritise potential disease-associated genes for further assessment. We evaluated new predisposition genes in exome sequencing data that we generated in 334 individuals with 27 other childhood cancers and in exome data from The Cancer Genome Atlas obtained from 7632 individuals with 28 adult cancers. FINDINGS: We identified constitutional cancer-predisposing mutations in 33 individuals with childhood cancer. The three identified genes with the strongest signal in the protein-truncating variant prioritisation analyses were TRIM28, FBXW7, and NYNRIN. 21 of 33 individuals had a mutation in TRIM28; there was a strong parent-of-origin effect, with all ten inherited mutations being maternally transmitted (p=0·00098). We also found a strong association with the rare epithelial subtype of Wilms tumour, with 14 of 16 tumours being epithelial or epithelial predominant. There were no TRIM28 mutations in individuals with other childhood or adult cancers. We identified truncating FBXW7 mutations in four individuals with Wilms tumour and a de-novo non-synonymous FBXW7 mutation in a child with a rhabdoid tumour. Biallelic truncating mutations in NYNRIN were identified in three individuals with Wilms tumour, which is highly unlikely to have occurred by chance (p<0·0001). Finally, we identified two de-novo KDM3B mutations, supporting the role of KDM3B as a childhood cancer predisposition gene. INTERPRETATION: The four new Wilms tumour predisposition genes identified-TRIM28, FBXW7, NYNRIN, and KDM3B-are involved in diverse biological processes and, together with the other 17 known Wilms tumour predisposition genes, account for about 10% of Wilms tumour cases. The overlap between these 21 constitutionally mutated predisposition genes and 20 genes somatically mutated in Wilms tumour is limited, consisting of only four genes. We recommend that all individuals with Wilms tumour should be offered genetic testing and particularly, those with epithelial Wilms tumour should be offered TRIM28 genetic testing. Only a third of the familial Wilms tumour clusters we analysed were attributable to known genes, indicating that further Wilms tumour predisposition factors await discovery. FUNDING: Wellcome Trust.


Asunto(s)
Genes del Tumor de Wilms , Tumor de Wilms/genética , Adolescente , Adulto , Niño , Preescolar , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Femenino , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Masculino , Persona de Mediana Edad , Mutación , Pronóstico , Proteína 28 que Contiene Motivos Tripartito/genética , Reino Unido/epidemiología , Secuenciación del Exoma , Tumor de Wilms/diagnóstico , Tumor de Wilms/mortalidad , Adulto Joven
19.
Wellcome Open Res ; 3: 46, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29900417

RESUMEN

Tatton-Brown-Rahman syndrome (TBRS; OMIM 615879), also known as the DNMT3A-overgrowth syndrome, is an overgrowth intellectual disability syndrome first described in 2014 with a report of 13 individuals with constitutive heterozygous DNMT3A variants. Here we have undertaken a detailed clinical study of 55 individuals with de novoDNMT3A variants, including the 13 previously reported individuals. An intellectual disability and overgrowth were reported in >80% of individuals with TBRS and were designated major clinical associations. Additional frequent clinical associations (reported in 20-80% individuals) included an evolving facial appearance with low-set, heavy, horizontal eyebrows and prominent upper central incisors; joint hypermobility (74%); obesity (weight ³2SD, 67%); hypotonia (54%); behavioural/psychiatric issues (most frequently autistic spectrum disorder, 51%); kyphoscoliosis (33%) and afebrile seizures (22%). One individual was diagnosed with acute myeloid leukaemia in teenage years. Based upon the results from this study, we present our current management for individuals with TBRS.

20.
Nat Genet ; 49(7): 1148-1151, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28553959

RESUMEN

Through exome sequencing, we identified six individuals with biallelic loss-of-function mutations in TRIP13. All six developed Wilms tumor. Constitutional mosaic aneuploidies, microcephaly, developmental delay and seizures, which are features of mosaic variegated aneuploidy (MVA) syndrome, were more variably present. Through functional studies, we show that TRIP13-mutant patient cells have no detectable TRIP13 and have substantial impairment of the spindle assembly checkpoint (SAC), leading to a high rate of chromosome missegregation. Accurate segregation, as well as SAC proficiency, is rescued by restoring TRIP13 function. Individuals with biallelic TRIP13 or BUB1B mutations have a high risk of embryonal tumors, and here we show that their cells display severe SAC impairment. MVA due to biallelic CEP57 mutations, or of unknown cause, is not associated with embryonal tumors and cells from these individuals show minimal SAC deficiency. These data provide insights into the complex relationships between aneuploidy and carcinogenesis.


Asunto(s)
Proteínas Portadoras/genética , Segregación Cromosómica/genética , Neoplasias Renales/genética , Puntos de Control de la Fase M del Ciclo Celular/genética , Tumor de Wilms/genética , ATPasas Asociadas con Actividades Celulares Diversas , Aneuploidia , Proteínas de Ciclo Celular/genética , Preescolar , ADN de Neoplasias/genética , Discapacidades del Desarrollo/genética , Femenino , Predisposición Genética a la Enfermedad , Humanos , Leucemia Mieloide Aguda/genética , Microcefalia/genética , Proteínas Asociadas a Microtúbulos/genética , Mosaicismo , Mutación , Neoplasias Primarias Múltiples/genética , Proteínas Nucleares/genética , Neoplasias Ováricas/genética , Proteínas Serina-Treonina Quinasas/genética , Estabilidad del ARN/genética , Convulsiones/genética , Tumor de Células de Sertoli-Leydig/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA