Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Base de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Neurology ; 100(4): e435-e442, 2023 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-36323522

RESUMEN

BACKGROUND AND OBJECTIVES: To determine the frequency and spectrum of complications of influenza infection in individuals with SCN1A-positive Dravet syndrome (SCN1A-DS). METHODS: Individuals with SCN1A-DS were identified in neurologists' care at 2 hospitals in Melbourne, Australia, with additional searches of EEG databases, the Victorian PAEDS FluCan influenza database, and the University of Melbourne Epilepsy Genetics Research Program database. Medical records were searched and families questioned to identify individuals who had an influenza infection; reported infections were confirmed by pathology report. For these individuals, we obtained baseline clinical characteristics and clinical details of the influenza infection. RESULTS: Twenty-one of 82 individuals (26%) had 24 documented influenza infections (17 influenza A and 7 influenza B) at age 0.5-25 years (median 4 years). All presented to hospital, 18/24 (75%) for status epilepticus or seizure exacerbations. Recovery was prompt in 18/24 (75%) infections, delayed but complete in 1/24 (4%) and incomplete in 5/24 (21%). One child died from influenza pneumonia, and long-term neurologic sequelae were seen with 4 infections. These individuals were poorly responsive after termination of status epilepticus. Brain imaging in 2 showed cerebral edema and 1 also having imaging features of laminar necrosis. All have ongoing neurologic deficits compared with their baseline, 1 having profound global impairment. DISCUSSION: Our data show that patients with SCN1A-DS are highly susceptible to neurologic complications during and severe sequelae after influenza infection, including moderate to severe persistent neurologic impairments and death. Safe administration of the seasonal influenza vaccine should be prioritized for this population.


Asunto(s)
Epilepsias Mioclónicas , Gripe Humana , Estado Epiléptico , Adolescente , Adulto , Niño , Preescolar , Humanos , Lactante , Adulto Joven , Epilepsias Mioclónicas/complicaciones , Epilepsias Mioclónicas/genética , Gripe Humana/complicaciones , Gripe Humana/epidemiología , Mutación , Canal de Sodio Activado por Voltaje NAV1.1/genética , Estado Epiléptico/complicaciones
2.
Nat Commun ; 12(1): 1482, 2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33674596

RESUMEN

Immune evasion is a hallmark of KRAS-driven cancers, but the underlying causes remain unresolved. Here, we use a mouse model of pancreatic ductal adenocarcinoma to inactivate KRAS by CRISPR-mediated genome editing. We demonstrate that at an advanced tumor stage, dependence on KRAS for tumor growth is reduced and is manifested in the suppression of antitumor immunity. KRAS-deficient cells retain the ability to form tumors in immunodeficient mice. However, they fail to evade the host immune system in syngeneic wild-type mice, triggering strong antitumor response. We uncover changes both in tumor cells and host immune cells attributable to oncogenic KRAS expression. We identify BRAF and MYC as key mediators of KRAS-driven tumor immune suppression and show that loss of BRAF effectively blocks tumor growth in mice. Applying our results to human PDAC we show that lowering KRAS activity is likewise associated with a more vigorous immune environment.


Asunto(s)
Evasión Inmune/fisiología , Modelos Genéticos , Neoplasias Pancreáticas/inmunología , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Edición Génica , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Conductos Pancreáticos/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas p21(ras)/inmunología , Transcriptoma , Neoplasias Pancreáticas
3.
Epilepsia ; 62(2): 358-370, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33475165

RESUMEN

OBJECTIVE: To study the epilepsy syndromes among the severe epilepsies of infancy and assess their incidence, etiologies, and outcomes. METHODS: A population-based cohort study was undertaken of severe epilepsies with onset before age 18 months in Victoria, Australia. Two epileptologists reviewed clinical features, seizure videos, and electroencephalograms to diagnose International League Against Epilepsy epilepsy syndromes. Incidence, etiologies, and outcomes at age 2 years were determined. RESULTS: Seventy-three of 114 (64%) infants fulfilled diagnostic criteria for epilepsy syndromes at presentation, and 16 (14%) had "variants" of epilepsy syndromes in which there was one missing or different feature, or where all classical features had not yet emerged. West syndrome (WS) and "WS-like" epilepsy (infantile spasms without hypsarrhythmia or modified hypsarrhythmia) were the most common syndromes, with a combined incidence of 32.7/100 000 live births/year. The incidence of epilepsy of infancy with migrating focal seizures (EIMFS) was 4.5/100 000 and of early infantile epileptic encephalopathy (EIEE) was 3.6/100 000. Structural etiologies were common in "WS-like" epilepsy (100%), unifocal epilepsy (83%), and WS (39%), whereas single gene disorders predominated in EIMFS, EIEE, and Dravet syndrome. Eighteen (16%) infants died before age 2 years. Development was delayed or borderline in 85 of 96 (89%) survivors, being severe-profound in 40 of 96 (42%). All infants with EIEE or EIMFS had severe-profound delay or were deceased, but only 19 of 64 (30%) infants with WS, "WS-like," or "unifocal epilepsy" had severe-profound delay, and only two of 64 (3%) were deceased. SIGNIFICANCE: Three quarters of severe epilepsies of infancy could be assigned an epilepsy syndrome or "variant syndrome" at presentation. In this era of genomic testing and advanced brain imaging, diagnosing epilepsy syndromes at presentation remains clinically useful for guiding etiologic investigation, initial treatment, and prognostication.


Asunto(s)
Discapacidades del Desarrollo/epidemiología , Epilepsias Mioclónicas/epidemiología , Espasmos Infantiles/epidemiología , Anticonvulsivantes/uso terapéutico , Preescolar , Estudios de Cohortes , Discapacidades del Desarrollo/etiología , Discapacidades del Desarrollo/fisiopatología , Progresión de la Enfermedad , Electroencefalografía , Epilepsias Mioclónicas/tratamiento farmacológico , Epilepsias Mioclónicas/etiología , Epilepsias Mioclónicas/fisiopatología , Síndromes Epilépticos/tratamiento farmacológico , Síndromes Epilépticos/epidemiología , Síndromes Epilépticos/etiología , Síndromes Epilépticos/fisiopatología , Femenino , Humanos , Incidencia , Lactante , Recién Nacido , Síndrome de Lennox-Gastaut/tratamiento farmacológico , Síndrome de Lennox-Gastaut/epidemiología , Síndrome de Lennox-Gastaut/etiología , Síndrome de Lennox-Gastaut/fisiopatología , Masculino , Malformaciones del Desarrollo Cortical/complicaciones , Malformaciones del Desarrollo Cortical/epidemiología , Malformaciones del Desarrollo Cortical/cirugía , Mortalidad , Índice de Severidad de la Enfermedad , Espasmos Infantiles/tratamiento farmacológico , Espasmos Infantiles/etiología , Espasmos Infantiles/fisiopatología , Victoria/epidemiología
4.
Neurology ; 94(20): e2148-e2167, 2020 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-32345733

RESUMEN

OBJECTIVE: Determining the genetic basis of speech disorders provides insight into the neurobiology of human communication. Despite intensive investigation over the past 2 decades, the etiology of most speech disorders in children remains unexplained. To test the hypothesis that speech disorders have a genetic etiology, we performed genetic analysis of children with severe speech disorder, specifically childhood apraxia of speech (CAS). METHODS: Precise phenotyping together with research genome or exome analysis were performed on children referred with a primary diagnosis of CAS. Gene coexpression and gene set enrichment analyses were conducted on high-confidence gene candidates. RESULTS: Thirty-four probands ascertained for CAS were studied. In 11/34 (32%) probands, we identified highly plausible pathogenic single nucleotide (n = 10; CDK13, EBF3, GNAO1, GNB1, DDX3X, MEIS2, POGZ, SETBP1, UPF2, ZNF142) or copy number (n = 1; 5q14.3q21.1 locus) variants in novel genes or loci for CAS. Testing of parental DNA was available for 9 probands and confirmed that the variants had arisen de novo. Eight genes encode proteins critical for regulation of gene transcription, and analyses of transcriptomic data found CAS-implicated genes were highly coexpressed in the developing human brain. CONCLUSION: We identify the likely genetic etiology in 11 patients with CAS and implicate 9 genes for the first time. We find that CAS is often a sporadic monogenic disorder, and highly genetically heterogeneous. Highly penetrant variants implicate shared pathways in broad transcriptional regulation, highlighting the key role of transcriptional regulation in normal speech development. CAS is a distinctive, socially debilitating clinical disorder, and understanding its molecular basis is the first step towards identifying precision medicine approaches.


Asunto(s)
Apraxias/genética , Trastornos del Habla/genética , Habla/fisiología , Factores de Transcripción/genética , Adolescente , Apraxias/diagnóstico , Apraxias/fisiopatología , Niño , Preescolar , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Estudios de Asociación Genética , Humanos , Masculino , Trastornos del Habla/diagnóstico , Trastornos del Habla/fisiopatología
5.
Eur J Paediatr Neurol ; 23(2): 280-287, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30744982

RESUMEN

BACKGROUND: Punctate white matter lesions (PWMLs) are small focal patches of increased signal intensity (SI) on T1- and decreased SI on T2-weighted magnetic resonance imaging (MRI). To date, there have been few reports of PWMLs in term born infants. OBJECTIVE: To identify associated diagnoses and factors predictive of clinical outcome in (near) term infants with PWMLs. METHODS: MRI studies and clinical records of (near) term infants, with PWMLs on MRI scans performed in two institutions in the first 28 postnatal days were reviewed. The PWMLs were classified according to their number, pattern and distribution. The medical records were examined to assess the associated diagnoses and determine the neurodevelopmental outcome at >12 months of age. Infants with congenital heart defect(s), those who had neonatal surgery, or those with perinatal arterial ischemic stroke were not eligible for the study. RESULTS: Forty-two (near) term infants with PWMLs were included. The major clinical association was perinatal asphyxia, present in 19/42 (45%). Ten (24%) had a history of seizures unrelated to asphyxia or a genetic diagnosis. Eleven (26%) had pathological genetic mutations. Other diagnoses, without seizures were identified in 2 (5%). The lesion load of PWMLs was high (>6) in 30/42 (71%). Evidence of irreversible white matter injury was present in 5 infants who had follow-up MRI performed between 18 and 24 months of age, because of clinical concerns. Five infants died and 37 had follow-up at a median age of 24 months. Neurodevelopmental outcome was poorest amongst 6 infants (16%) whose PWMLs occurred in the setting of a genetic disorder. CONCLUSION: PWMLs in (near) term infants represent white matter injury that may evolve into gliosis and/or white matter loss. Infants with PWMLs in the setting of a genetic disorder appeared at most risk of a poor outcome.


Asunto(s)
Encéfalo/patología , Enfermedades del Recién Nacido/patología , Sustancia Blanca/patología , Encéfalo/diagnóstico por imagen , Femenino , Humanos , Recién Nacido , Enfermedades del Recién Nacido/diagnóstico por imagen , Imagen por Resonancia Magnética , Masculino , Sustancia Blanca/diagnóstico por imagen
6.
Int J Cancer ; 142(1): 156-164, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28906000

RESUMEN

Tyrosine kinase inhibitors are effective treatments for cancers. Knowing the specific kinase mutants that drive the underlying cancers predict therapeutic response to these inhibitors. Thus, the current protocol for personalized cancer therapy involves genotyping tumors in search of various driver mutations and subsequently individualizing the tyrosine kinase inhibitor to the patients whose tumors express the corresponding driver mutant. While this approach works when known driver mutations are found, its limitation is the dependence on driver mutations as predictors for response. To complement the genotype approach, we hypothesize that a phosphoarray platform is equally capable of personalizing kinase inhibitor therapy. We selected head and neck squamous cell carcinoma as the cancer model to test our hypothesis. Using the receptor tyrosine kinase phosphoarray, we identified the phosphorylation profiles of 49 different tyrosine kinase receptors in five different head and neck cancer cell lines. Based on these results, we tested the cell line response to the corresponding kinase inhibitor therapy. We found that this phosphoarray accurately informed the kinase inhibitor response profile of the cell lines. Next, we determined the phosphorylation profiles of 39 head and neck cancer patient derived xenografts. We found that absent phosphorylated EGFR signal predicted primary resistance to cetuximab treatment in the xenografts without phosphorylated ErbB2. Meanwhile, absent ErbB2 signaling in the xenografts with phosphorylated EGFR is associated with a higher likelihood of response to cetuximab. In summary, the phosphoarray technology has the potential to become a new diagnostic platform for personalized cancer therapy.


Asunto(s)
Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Ensayos Analíticos de Alto Rendimiento/métodos , Medicina de Precisión/métodos , Proteínas Tirosina Quinasas/análisis , Animales , Antineoplásicos/farmacología , Cetuximab/farmacología , Resistencia a Antineoplásicos/fisiología , Humanos , Ratones , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Oncotarget ; 8(11): 17995-18009, 2017 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-28152508

RESUMEN

KRAS is the most commonly mutated oncogene, frequently associated with some of the deadliest forms of cancer. However, the need for potent and specific KRAS inhibitors remains unmet. Here, we evaluated the effects of selected cytotoxic agents on oncogenic KRAS signaling and drug response. The data provided new insights into the functional interaction between the KRAS and MYC pathways and revealed key differences between WT and mutant KRAS expressing cells. Systematic investigation of non-small cell lung cancer cell lines revealed that KRAS mutation can paradoxically increase the sensitivity of cells to cytotoxic agents. We identify MYC as a key regulator of the cellular stress responses and tumor cell viability as MYC expression was suppressed in drug-sensitive but not resistant cells. Furthermore, this suppression was driven by hyperactive KRAS/MAPK signaling. Our findings support a direct link between MYC and cancer cell viability, and raise the possibility that inactivation of MYC may be an effective therapeutic strategy for KRAS mutant tumors across various cancer types.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Resistencia a Antineoplásicos/genética , Neoplasias Pulmonares/genética , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Antineoplásicos/farmacología , Western Blotting , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Citotoxinas/farmacología , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Neoplasias Pulmonares/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Transducción de Señal/efectos de los fármacos
8.
Theranostics ; 6(6): 808-16, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27162551

RESUMEN

Lack of absolute selectivity against cancer cells is a major limitation for current cancer therapies. In the previous study, we developed a prodrug strategy for selective cancer therapy using a masked cytotoxic agent puromycin [Boc-Lys(Ac)-Puromycin], which can be sequentially activated by histone deacetylases (HDACs) and cathepsin L (CTSL) to kill cancer cells expressing high levels of both enzymes. Despite the promise as a selective cancer therapy, its requirement of relatively high dosage could be a potential issue in the clinical setting. To address this issue, we aimed to further improve the overall efficacy of our prodrug strategy. Since the proteolytic cleavage by CTSL is the rate-limiting step for the drug activation, we sought to improve the substrate structure for CTSL activity by modifying the α-amino protecting group of lysine. Here we show that protection with Fmoc [Fmoc-Lys(Ac)-Puromycin] exhibits a marked improvement in overall anticancer efficacy compared to the original Boc-Lys(Ac)-Puromycin and this is mainly due to the highly efficient cellular uptake besides its improved substrate structure. Furthermore, to address a concern that the improved drug efficacy might direct high toxicity to the normal cells, we confirmed that Fmoc-Lys(Ac)-Puromycin still retains excellent cancer selectivity in vitro and no obvious systemic off-target toxicity in vivo. Thus our preclinical evaluation data presented here demonstrate that the Fmoc-Lys(Ac)-Puromycin exhibits substantially improved anticancer efficacy, further supporting our approach for the selective cancer therapy.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Catepsina L/metabolismo , Histona Desacetilasas/metabolismo , Profármacos/síntesis química , Profármacos/farmacología , Animales , Antineoplásicos/efectos adversos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Femenino , Xenoinjertos , Humanos , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Profármacos/efectos adversos , Resultado del Tratamiento
9.
Mol Cancer Res ; 14(8): 740-52, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27216155

RESUMEN

UNLABELLED: EGFR is a popular therapeutic target for many cancers. EGFR inhibitors have been tested in children with refractory neuroblastoma. Interestingly, partial response or stable disease was observed in a few neuroblastoma patients. As EGFR mutations are biomarkers for response to anti-EGFR drugs, primary neuroblastoma tumors and cell lines were screened for mutations. A novel EGFR extracellular domain deletion mutant, EGFRΔ768, was discovered and the biologic and biochemical properties of this mutant were characterized and compared with wild-type and EGFRvIII receptors. EGFRΔ768 was found to be constitutively active and localized to the cell surface. Its expression conferred resistance to etoposide and drove proliferation as well as invasion of cancer cells. While EGFRΔ768 had similarity to EGFRvIII, its biologic and biochemical properties were distinctly different from both the EGFRvIII and wild-type receptors. Even though erlotinib inhibited EGFRΔ768, its effect on the mutant was not as strong as that on wild-type EGFR and EGFRvIII. In addition, downstream signaling of EGFRΔ768 was different from that of the wild-type receptor. In conclusion, this is the first study to demonstrate that neuroblastoma express not only EGFRvIII, but also a novel EGFR extracellular domain deletion mutant, EGFRΔ768. The EGFRΔ768 also possesses distinct biologic and biochemical properties which might have therapeutic implications for neuroblastoma as well as other tumors expressing this novel mutant. IMPLICATIONS: Neuroblastoma expressed a novel EGFR mutant which possesses distinct biologic and biochemical properties that might have therapeutic implications. Mol Cancer Res; 14(8); 740-52. ©2016 AACR.


Asunto(s)
Receptores ErbB/genética , Neuroblastoma/genética , Secuencia de Aminoácidos , Línea Celular Tumoral , Humanos , Mutación , Fosforilación , Transducción de Señal , Transfección
10.
J Cell Biochem ; 117(2): 334-43, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26138431

RESUMEN

Oncogenic kinase Aurora A (AURKA) has been found to be overexpresed in several tumors including colorectal, breast, and hematological cancers. Overexpression of AURKA induces centrosome amplification and aneuploidy and it is related with cancer progression and poor prognosis. Here we show that AURKA phosphorylates in vitro the transcripcional co-repressor Ski on aminoacids Ser326 and Ser383. Phosphorylations on these aminoacids decreased Ski protein half-life. Reduced levels of Ski resulted in centrosomes amplification and multipolar spindles formation, same as AURKA overexpressing cells. Importantly, overexpression of Ski wild type, but not S326D and S383D mutants inhibited centrosome amplification and cellular transformation induced by AURKA. Altogether, these results suggest that the Ski protein is a target in the transformation pathway mediated by the AURKA oncogene.


Asunto(s)
Aurora Quinasa A/metabolismo , Transformación Celular Neoplásica/metabolismo , Proteínas de Unión al ADN/fisiología , Proteínas Proto-Oncogénicas/fisiología , Secuencia de Aminoácidos , Animales , Centrosoma/metabolismo , Expresión Génica , Células HEK293 , Humanos , Células MCF-7 , Ratones , Datos de Secuencia Molecular , Células 3T3 NIH , Fosforilación , Procesamiento Proteico-Postraduccional , Huso Acromático/metabolismo
11.
Oncotarget ; 6(18): 15814-27, 2015 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-26158412

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive, metastatic disease with limited treatment options. Factors contributing to the metastatic predisposition and therapy resistance in pancreatic cancer are not well understood. Here, we used a mouse model of KRAS-driven pancreatic carcinogenesis to define distinct subtypes of PDAC metastasis: epithelial, mesenchymal and quasi-mesenchymal. We examined pro-survival signals in these cells and the therapeutic response differences between them. Our data indicate that the initiation and maintenance of the transformed state are separable, and that KRAS dependency is not a fundamental constant of KRAS-initiated tumors. Moreover, some cancer cells can shuttle between the KRAS dependent (drug-sensitive) and independent (drug-tolerant) states and thus escape extinction. We further demonstrate that inhibition of KRAS signaling alone via co-targeting the MAPK and PI3K pathways fails to induce extensive tumor cell death and, therefore, has limited efficacy against PDAC. However, the addition of histone deacetylase (HDAC) inhibitors greatly improves outcomes, reduces the self-renewal of cancer cells, and blocks cancer metastasis in vivo. Our results suggest that targeting HDACs in combination with KRAS or its effector pathways provides an effective strategy for the treatment of PDAC.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/farmacología , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Neoplasias Pancreáticas/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Animales , Carcinoma Ductal Pancreático/enzimología , Carcinoma Ductal Pancreático/genética , Línea Celular Tumoral , Resistencia a Antineoplásicos , Genes ras , Inhibidores de Histona Desacetilasas/administración & dosificación , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Desnudos , Mutación , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/genética , Inhibidores de Proteínas Quinasas/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Epilepsia ; 56(7): 1071-80, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25982755

RESUMEN

OBJECTIVE: We evaluated seizure outcome in a large cohort of familial neonatal seizures (FNS), and examined phenotypic overlap with different molecular lesions. METHODS: Detailed clinical data were collected from 36 families comprising two or more individuals with neonatal seizures. The seizure course and occurrence of seizures later in life were analyzed. Families were screened for KCNQ2, KCNQ3, SCN2A, and PRRT2 mutations, and linkage studies were performed in mutation-negative families to exclude known loci. RESULTS: Thirty-three families fulfilled clinical criteria for benign familial neonatal epilepsy (BFNE); 27 of these families had KCNQ2 mutations, one had a KCNQ3 mutation, and two had SCN2A mutations. Seizures persisting after age 6 months were reported in 31% of individuals with KCNQ2 mutations; later seizures were associated with frequent neonatal seizures. Linkage mapping in two mutation-negative BFNE families excluded linkage to KCNQ2, KCNQ3, and SCN2A, but linkage to KCNQ2 could not be excluded in the third mutation-negative BFNE family. The three remaining families did not fulfill criteria of BFNE due to developmental delay or intellectual disability; a molecular lesion was identified in two; the other family remains unsolved. SIGNIFICANCE: Most families in our cohort of familial neonatal seizures fulfill criteria for BFNE; the molecular cause was identified in 91%. Most had KCNQ2 mutations, but two families had SCN2A mutations, which are normally associated with a mixed picture of neonatal and infantile onset seizures. Seizures later in life are more common in BFNE than previously reported and are associated with a greater number of seizures in the neonatal period. Linkage studies in two families excluded known loci, suggesting a further gene is involved in BFNE.


Asunto(s)
Epilepsia Benigna Neonatal/diagnóstico , Epilepsia Benigna Neonatal/genética , Estudios de Cohortes , Femenino , Humanos , Recién Nacido , Canal de Potasio KCNQ2 , Masculino , Linaje , Convulsiones , Resultado del Tratamiento
13.
Proc Natl Acad Sci U S A ; 111(9): 3466-71, 2014 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-24550494

RESUMEN

Pancreatic cancer is one of the deadliest human malignancies. A striking feature of pancreatic cancer is that activating Kras mutations are found in ∼90% of cases. However, apart from a restricted population of cells expressing pancreatic and duodenal homeobox 1 (PDX1), most pancreatic cells are refractory to Kras-driven transformation. In the present study, we sought to determine which subsets of PDX1+ cells may be responsible for tumor growth. Using the Lox-Stop-Lox-KrasG12D genetic mouse model of pancreatic carcinogenesis, we isolated a population of KrasG12D-expressing PDX1+ cells with an inherent capacity to metastasize. This population of cells bears the surface phenotype of EpCAM+CD24+CD44+CD133-SCA1- and is closer in its properties to stem-like cells than to more mature cell types. We further demonstrate that the tumorigenic capacity of PDX1+ cells is limited, becoming progressively lost as the cells acquire a mature phenotype. These data are consistent with the hypothesis that the adult pancreas harbors a dormant progenitor cell population that is capable of initiating tumor growth under conditions of oncogenic stimulation. We present evidence that constitutive activation of the mitogen-activated protein kinase (MAPK/ERK) signaling and stabilization of the MYC protein are the two main driving forces behind the development of pancreatic cancer cells with stem-cell-like properties and high metastatic potential. Our results suggest that pancreatic cells bearing Kras mutation can be induced to differentiate into quasi-normal cells with suppressed tumorigenicity by selective inhibition of the MAPK/ERK/MYC signaling cascade.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Neoplasias Pulmonares/secundario , Sistema de Señalización de MAP Quinasas/fisiología , Metástasis de la Neoplasia/fisiopatología , Neoplasias Pancreáticas/fisiopatología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Transactivadores/metabolismo , Animales , Western Blotting , Citometría de Flujo , Neoplasias Pulmonares/patología , Ratones , Ratones Noqueados , Ratones Desnudos , Mutación Missense/genética , Neoplasias Pancreáticas/metabolismo , Reacción en Cadena de la Polimerasa , Proteína p53 Supresora de Tumor/genética
14.
Nat Commun ; 4: 2735, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24193185

RESUMEN

Eradication of cancer cells while minimizing damage to healthy cells is a primary goal of cancer therapy. Highly selective drugs are urgently needed. Here we demonstrate a new prodrug strategy for selective cancer therapy that utilizes increased histone deacetylase (HDAC) and tumour-associated protease activities produced in malignant cancer cells. By coupling an acetylated lysine group to puromycin, a masked cytotoxic agent is created, which is serially activated by HDAC and an endogenous protease cathepsin L (CTSL) that remove the acetyl group first and then the unacetylated lysine group liberating puromycin. The agent selectively kills human cancer cell lines with high HDAC and CTSL activities. In vivo studies confirm tumour growth inhibition in prodrug-treated mice bearing human cancer xenografts. This cancer-selective cleavage of the masking group is a promising strategy for the next generation of anticancer drug development that could be applied to many other cytotoxic agents.


Asunto(s)
Antineoplásicos/uso terapéutico , Catepsina L/metabolismo , Histona Desacetilasas/metabolismo , Neoplasias Experimentales/tratamiento farmacológico , Profármacos , Puromicina/uso terapéutico , Animales , Línea Celular Tumoral , Supervivencia Celular , Regulación Enzimológica de la Expresión Génica/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Ratones , Neoplasias Experimentales/enzimología , Puromicina/química , Puromicina/metabolismo
15.
J Paediatr Child Health ; 49(9): 716-24, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23731025

RESUMEN

AIM: Despite advances in medical investigation, many children with neurological conditions remain without a diagnosis, although a genetic aetiology is often suspected. Chromosomal microarray (CMA) screens for copy number variants (CNVs) and long continuous stretches of homozygosity (LCSH) and may further enhance diagnostic yield. Although recent studies have identified pathogenic CNVs in intellectual disability, autism and epilepsy, the utility of CMA testing in a broader cohort of children with neurologic disorders has not been reported. METHODS: Two hundred fifteen patients with neurological conditions of unknown aetiology were seen over a 6-month period and were prospectively tested by CMA using high-resolution single nucleotide polymorphism (SNP) microarrays (Illumina HumanCytoSNP-12 v2.1 or Affymetrix 2.7M). RESULTS: Thirty of 215 (14%) patients tested had an abnormal CMA. Twenty-nine had CNVs (13%) and one (0.5%) a clinically significant stretch of homozygosity. Twenty (9.3%) had a CMA finding considered to be pathogenic or involved in susceptibility to the condition of interest, and 10 (4.7%) had findings of unknown significance. Their phenotypes included infantile spasms and other epilepsies, neuromuscular conditions, ataxia, movement disorders, microcephaly and malformations of cortical development. At least one third of patients did not meet national funding criteria for CMA at the time of presentation. CONCLUSIONS: CMA detected clinically significant abnormalities in a broad range of neurologic phenotypes of unknown aetiology. This test should be considered a first-tier investigation of children with neurologic disorders in whom the initial clinical assessment does not indicate a likely aetiology, especially those with severe epilepsies and neurologically abnormal neonates.


Asunto(s)
Variaciones en el Número de Copia de ADN , Predisposición Genética a la Enfermedad , Enfermedades del Sistema Nervioso/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Polimorfismo de Nucleótido Simple , Niño , Preescolar , Homocigoto , Humanos , Lactante , Recién Nacido , Fenotipo , Estudios Prospectivos
16.
BMC Biochem ; 14: 10, 2013 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-23566155

RESUMEN

BACKGROUND: The androgen receptor (AR) is a member of the nuclear receptor (NR) superfamily of ligand-inducible DNA transcription factors, and is the major mediator of male sexual development, prostate growth and the pathogenesis of prostate cancer. Cell and gene specific regulation by the AR is determined by availability of and interaction with sets of key accessory cofactors. Ski-interacting protein (SKIP; SNW1, NCOA62) is a cofactor shown to interact with several NRs and a diverse range of other transcription factors. Interestingly, SKIP as part of the spliceosome is thought to link mRNA splicing with transcription. SKIP has not been previously shown to interact with the AR. RESULTS: The aim of this study was to investigate whether SKIP interacts with the AR and modulates AR-dependent transcription. Here, we show by co-immunoprecipitation experiments that SKIP is in a complex with the AR. Moreover, SKIP increased 5α-dihydrotestosterone (DHT) induced N-terminal/C-terminal AR interaction from 12-fold to almost 300-fold in a two-hybrid assay, and enhanced AR ligand-independent AF-1 transactivation. SKIP augmented ligand- and AR-dependent transactivation in PC3 prostate cancer cells. Live-cell imaging revealed a fast (half-time=129 s) translocation of AR from the cytoplasm to the nucleus upon DHT-stimulation. Förster resonance energy transfer (FRET) experiments suggest a direct AR-SKIP interaction in the nucleus upon translocation. CONCLUSIONS: Our results suggest that SKIP interacts with AR in the nucleus and enhances AR-dependent transactivation and N/C-interaction supporting a role for SKIP as an AR co-factor.


Asunto(s)
Coactivadores de Receptor Nuclear/metabolismo , Receptores Androgénicos/metabolismo , Animales , Células COS , Línea Celular Tumoral , Núcleo Celular/metabolismo , Chlorocebus aethiops , Dihidrotestosterona/farmacología , Transferencia Resonante de Energía de Fluorescencia , Genes Reporteros , Células HEK293 , Humanos , Inmunoprecipitación , Masculino , Coactivadores de Receptor Nuclear/genética , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Receptores Androgénicos/química , Receptores Androgénicos/genética , Activación Transcripcional
17.
BMC Infect Dis ; 12: 105, 2012 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-22545865

RESUMEN

BACKGROUND: Recurrent benign 6th nerve palsy in the paediatric age group is uncommon, but has been described following viral and bacterial infections. It has also been temporally associated with immunization, but has not been previously described following two different live attenuated vaccines. CASE PRESENTATION: A case is presented of a 12 month old Caucasian boy with recurrent benign 6th nerve palsy following measles-mumps-rubella and varicella vaccines, given on separate occasions with complete recovery following each episode. No alternate underlying etiology was identified despite extensive investigations and review. CONCLUSIONS: The majority of benign 6th nerve palsies do not have a sinister cause and have an excellent prognosis, with recovery expected in most cases. The exact pathophysiology is unknown, although hypotheses including autoimmune mechanisms and direct viral invasion could explain the pathophysiology behind immunization related nerve palsies. It is important to rule out other aetiologies with thorough history, physical examination and investigations. There is limited information in the literature regarding the safety of a repeat dose of a live vaccine in this setting. Future immunizations should be considered on a case-by-case basis.


Asunto(s)
Enfermedades del Nervio Abducens/inducido químicamente , Enfermedades del Nervio Abducens/diagnóstico , Vacuna contra la Varicela/efectos adversos , Vacuna contra el Sarampión-Parotiditis-Rubéola/efectos adversos , Vacunación/efectos adversos , Enfermedades del Nervio Abducens/patología , Vacuna contra la Varicela/administración & dosificación , Humanos , Lactante , Masculino , Vacuna contra el Sarampión-Parotiditis-Rubéola/administración & dosificación , Recurrencia , Vacunación/métodos , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/efectos adversos
18.
J Cell Physiol ; 227(1): 278-87, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21412778

RESUMEN

Ski is a transcriptional regulator that has been considered an oncoprotein given its ability to induce oncogenic transformation in avian model systems. However, studies in mouse and in some human tumor cells have also indicated a tumor suppressor activity for this protein. We found that Ski-/- mouse embryo fibroblasts exhibit high levels of genome instability, namely aneuploidy, consistent with a tumor suppressor function for Ski. Time-lapse microscopy revealed lagging chromosomes and chromatin/chromosome bridges as the major cause of micronuclei (MN) formation and the subsequent aneuploidy. Although these cells arrested in mitosis after treatment with spindle disrupting drugs and exhibited a delayed metaphase/anaphase transition, spindle assembly checkpoint (SAC) was not sufficient to prevent chromosome missegregation, consistent with a weakened SAC. Our in vivo analysis also showed dynamic metaphase plate rearrangements with switches in polarity in cells arrested in metaphase. Importantly, after ectopic expression of Ski the cells that displayed this metaphase arrest died directly during metaphase or after aberrant cell division, relating SAC activation and mitotic cell death. This increased susceptibility to undergo mitosis-associated cell death reduced the number of MN-containing cells. The presented data support a new role for Ski in the mitotic process and in maintenance of genetic stability, providing insights into the mechanism of tumor suppression mediated by this protein.


Asunto(s)
Transformación Celular Neoplásica/genética , Inestabilidad Cromosómica/genética , Proteínas de Unión al ADN/genética , Fibroblastos/patología , Proteínas Proto-Oncogénicas/genética , Animales , Separación Celular , Células Cultivadas , Embrión de Mamíferos , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Immunoblotting , Ratones , Ratones Noqueados , Mitosis/genética , Transcripción Genética
19.
J Pediatr ; 160(1): 165-8.e1, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21982304

RESUMEN

We describe 5 preterm and 3 term infants who presented with seizures during rotavirus infection within 6 weeks after birth. Six of these infants developed late-onset cystic periventricular leukomalacia. Four of the preterm infants had neurodevelopmental delay, and 4 (near) term infants had normal early outcome.


Asunto(s)
Leucomalacia Periventricular/virología , Infecciones por Rotavirus/complicaciones , Femenino , Humanos , Recién Nacido , Masculino
20.
Biochem Biophys Res Commun ; 409(3): 539-43, 2011 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-21600873

RESUMEN

Ski is a negative regulator of the transforming growth factor-ß and other signalling pathways. The absence of SKI in mouse fibroblasts leads to chromosome segregation defects and genomic instability, suggesting a role for Ski during mitosis. At this stage, Ski is phosphorylated but to date little is known about the kinases involved in this process. Here, we show that Aurora A kinase is able to phosphorylate Ski in vitro. In vivo, Aurora A and Ski co-localized at the centrosomes and co-immunoprecipitated. Conversely, a C-terminal truncation mutant of Ski (SkiΔ491-728) lacking a coiled-coil domain, displayed decreased centrosomal localization. This mutant no longer co-immunoprecipitated with Aurora-A in vivo, but was still phosphorylated in vitro, indicating that the Ski-Aurora A interaction takes place at the centrosomes. These data identify Ski as a novel target of Aurora A and contribute to an understanding of the role of these proteins in the mitotic process.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Mitosis , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Aurora Quinasa A , Aurora Quinasas , Línea Celular Tumoral , Centrómero/metabolismo , Centrosoma/metabolismo , Proteínas de Unión al ADN/genética , Células HEK293 , Humanos , Inmunoprecipitación , Ratones , Mutación , Fosforilación , Proteínas Proto-Oncogénicas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA