Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 525(7567): 109-13, 2015 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-26258302

RESUMEN

Mitral valve prolapse (MVP) is a common cardiac valve disease that affects nearly 1 in 40 individuals. It can manifest as mitral regurgitation and is the leading indication for mitral valve surgery. Despite a clear heritable component, the genetic aetiology leading to non-syndromic MVP has remained elusive. Four affected individuals from a large multigenerational family segregating non-syndromic MVP underwent capture sequencing of the linked interval on chromosome 11. We report a missense mutation in the DCHS1 gene, the human homologue of the Drosophila cell polarity gene dachsous (ds), that segregates with MVP in the family. Morpholino knockdown of the zebrafish homologue dachsous1b resulted in a cardiac atrioventricular canal defect that could be rescued by wild-type human DCHS1, but not by DCHS1 messenger RNA with the familial mutation. Further genetic studies identified two additional families in which a second deleterious DCHS1 mutation segregates with MVP. Both DCHS1 mutations reduce protein stability as demonstrated in zebrafish, cultured cells and, notably, in mitral valve interstitial cells (MVICs) obtained during mitral valve repair surgery of a proband. Dchs1(+/-) mice had prolapse of thickened mitral leaflets, which could be traced back to developmental errors in valve morphogenesis. DCHS1 deficiency in MVP patient MVICs, as well as in Dchs1(+/-) mouse MVICs, result in altered migration and cellular patterning, supporting these processes as aetiological underpinnings for the disease. Understanding the role of DCHS1 in mitral valve development and MVP pathogenesis holds potential for therapeutic insights for this very common disease.


Asunto(s)
Cadherinas/genética , Cadherinas/metabolismo , Prolapso de la Válvula Mitral/genética , Prolapso de la Válvula Mitral/patología , Mutación/genética , Animales , Tipificación del Cuerpo/genética , Proteínas Relacionadas con las Cadherinas , Cadherinas/deficiencia , Movimiento Celular/genética , Cromosomas Humanos Par 11/genética , Femenino , Humanos , Masculino , Ratones , Válvula Mitral/anomalías , Válvula Mitral/embriología , Válvula Mitral/patología , Válvula Mitral/cirugía , Linaje , Fenotipo , Estabilidad Proteica , ARN Mensajero/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
3.
Hum Mol Genet ; 20(21): 4093-101, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21821670

RESUMEN

Recent studies emphasize the importance of mRNA splicing in human genetic disease, as 20-30% of all disease-causing mutations are predicted to result in mRNA splicing defects. The plasticity of the mRNA splicing reaction has made these mutations attractive candidates for the development of therapeutics. Familial dysautonomia (FD) is a severe neurodegenerative disorder, and all patients have an intronic IVS20+6T>C splice site mutation in the IKBKAP gene, which results in tissue-specific skipping of exon 20 and a corresponding reduction in ikappaB kinase complex associated protein (IKAP) levels. We created transgenic mouse lines using a human IKBKAP bacterial artificial chromosome (BAC) into which we inserted the IKBKAP splice mutation (FD BAC) and have shown that the transgenic mice exhibit the same tissue-specific aberrant splicing patterns as seen in FD patients. We have previously demonstrated that the plant cytokinin kinetin can significantly improve the production of wild-type IKBKAP transcripts in FD lymphoblast cell lines by improving exon inclusion. In this study, we tested the ability of kinetin to alter IKBKAP splicing in the transgenic mice carrying the FD BAC and show that it corrects IKBKAP splicing in all major tissues assayed, including the brain. The amount of wild-type IKBKAP mRNA and IKAP protein was significantly higher in the kinetin-treated mice. These exciting results prove that treatment of FD, as well as other mechanistically related splicing disorders, with kinetin holds great promise as a potential therapeutic aimed at increasing normal protein levels, which may, in turn, slow disease progression.


Asunto(s)
Empalme Alternativo/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Suplementos Dietéticos , Cinetina/farmacología , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Células Cultivadas , Dieta , Relación Dosis-Respuesta a Droga , Péptidos y Proteínas de Señalización Intracelular , Cinetina/administración & dosificación , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/metabolismo
4.
Artículo en Inglés | MEDLINE | ID: mdl-37377441

RESUMEN

INTRODUCTION: Leadership skills are essential for a successful career in medical research but are often not formally taught. To address these gaps, we designed a leadership development program for early-stage investigators. METHODS: A 9-month virtual program with monthly 2-hour interactive sessions was designed, covering topics such as Leadership in Research, Mentoring, Building Diverse and Inclusive Teams, Managing Conflict, Influencing without Authority, Grant Administration, and Management. An anonymized survey was sent to participants before and after completion of the program, and the results were compared using the chi-squared test. RESULTS: Over a 2-year period, we selected two cohorts of 41 and 46 participants, respectively. After completion of the program, 92% of survey respondents indicated that the program met their expectations and 74% had made use of skills they learned. Participants enjoyed meeting new people and discussing common challenges. There was an increase in participants' perceived understanding of personal leadership qualities, mentoring, communication, conflict resolution, grant management, and collaboration with industry (P < .05). DISCUSSION: A leadership development program for early-stage investigators led to a significant increase in participants' perceived understanding of personal leadership qualities and competencies. It also offered participants the opportunity to meet other researchers in the institution and discuss common challenges.

5.
Pediatr Res ; 70(5): 480-3, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21775922

RESUMEN

Familial dysautonomia (FD) is caused by an intronic splice mutation in the IKBKAP gene that leads to partial skipping of exon 20 and tissue-specific reduction in I-κ-B kinase complex-associated protein/elongation protein 1 (IKAP/ELP-1) expression. Kinetin (6-furfurylaminopurine) has been shown to improve splicing and increase WT IKBKAP mRNA and IKAP protein expression in FD cell lines and carriers. To determine whether oral kinetin treatment could alter mRNA splicing in FD subjects and was tolerable, we administered kinetin to eight FD individuals homozygous for the splice mutation. Subjects received 23.5 mg/Kg/d for 28 d. An increase in WT IKBKAP mRNA expression in leukocytes was noted after 8 d in six of eight individuals; after 28 d, the mean increase compared with baseline was significant (p = 0.002). We have demonstrated that kinetin is tolerable in this medically fragile population. Not only did kinetin produce the desired effect on splicing in FD patients but also that effect seems to improve with time despite lack of dose change. This is the first report of a drug that produces in vivo mRNA splicing changes in individuals with FD and supports future long-term trials to determine whether kinetin will prove therapeutic in FD patients.


Asunto(s)
Proteínas Portadoras/metabolismo , Disautonomía Familiar/fisiopatología , Regulación de la Expresión Génica/efectos de los fármacos , Cinetina/farmacología , Empalme del ARN/efectos de los fármacos , ARN Mensajero/metabolismo , Administración Oral , Adulto , Análisis de Varianza , Área Bajo la Curva , Proteínas Portadoras/genética , Disautonomía Familiar/genética , Femenino , Humanos , Cinetina/administración & dosificación , Cinetina/sangre , Cinetina/farmacocinética , Masculino , New York , Empalme del ARN/fisiología , Factores de Elongación Transcripcional
6.
Pediatr Res ; 65(3): 341-6, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19033881

RESUMEN

Familial dysautonomia (FD) is caused by an intronic splice mutation in the IkappaB kinase-associated protein gene (IKBKAP) that leads to partial skipping of exon 20 and tissue-specific reduction of IkappaB kinase-associated protein/elongator protein 1 (IKAP/ELP-1 protein). Kinetin increases IKBKAP mRNA and protein expression in FD cell lines. To determine whether oral kinetin alters IKBKAP splicing in vivo, we administered kinetin to 29 healthy carriers of the major FD mutation for 8 d. Adverse effects, kinetin, and IKBKAP mRNA levels were monitored. In the highest dosing cohorts (23.5 mg/kg/d), the target plasma kinetin level was achieved in 91% of subjects at 2 h. After 8 d, IKBKAP mRNA expression in leukocytes increased as kinetin levels increased. There is a linear association between log plasma kinetin level and corresponding log change from baseline in IKBKAP mRNA expression that allows estimation of IKBKAP mRNA levels because of kinetin ingestion. Adverse effects were transient and mild. This is the first report of in vivo IKBKAP splicing modification and strongly suggests kinetin's therapeutic potential in FD and perhaps in other splicing disorders. Furthermore, our findings support our hypothesis that treatments, which target a particular splicing mutation, can be successfully developed.


Asunto(s)
Empalme Alternativo/genética , Proteínas Portadoras/genética , Disautonomía Familiar/genética , Regulación de la Expresión Génica/efectos de los fármacos , Heterocigoto , Cinetina/farmacología , ARN Mensajero/metabolismo , Adulto , Empalme Alternativo/efectos de los fármacos , Proteínas Portadoras/metabolismo , Relación Dosis-Respuesta a Droga , Disautonomía Familiar/tratamiento farmacológico , Femenino , Humanos , Cinetina/sangre , Cinetina/farmacocinética , Masculino , Mutación/genética , ARN Mensajero/genética , Estadísticas no Paramétricas , Factores de Elongación Transcripcional
7.
Sci Transl Med ; 11(493)2019 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-31118289

RESUMEN

Mitral valve prolapse (MVP) affects 1 in 40 people and is the most common indication for mitral valve surgery. MVP can cause arrhythmias, heart failure, and sudden cardiac death, and to date, the causes of this disease are poorly understood. We now demonstrate that defects in primary cilia genes and their regulated pathways can cause MVP in familial and sporadic nonsyndromic MVP cases. Our expression studies and genetic ablation experiments confirmed a role for primary cilia in regulating ECM deposition during cardiac development. Loss of primary cilia during development resulted in progressive myxomatous degeneration and profound mitral valve pathology in the adult setting. Analysis of a large family with inherited, autosomal dominant nonsyndromic MVP identified a deleterious missense mutation in a cilia gene, DZIP1 A mouse model harboring this variant confirmed the pathogenicity of this mutation and revealed impaired ciliogenesis during development, which progressed to adult myxomatous valve disease and functional MVP. Relevance of primary cilia in common forms of MVP was tested using pathway enrichment in a large population of patients with MVP and controls from previously generated genome-wide association studies (GWAS), which confirmed the involvement of primary cilia genes in MVP. Together, our studies establish a developmental basis for MVP through altered cilia-dependent regulation of ECM and suggest that defects in primary cilia genes can be causative to disease phenotype in some patients with MVP.


Asunto(s)
Cilios/patología , Prolapso de la Válvula Mitral/etiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Secuencia de Bases , Matriz Extracelular/metabolismo , Femenino , Estudio de Asociación del Genoma Completo , Válvulas Cardíacas/diagnóstico por imagen , Válvulas Cardíacas/crecimiento & desarrollo , Humanos , Masculino , Ratones Noqueados , Prolapso de la Válvula Mitral/diagnóstico por imagen , Prolapso de la Válvula Mitral/genética , Morfogénesis , Linaje , Factores de Tiempo , Proteínas Supresoras de Tumor/metabolismo
8.
J Mol Med (Berl) ; 85(2): 149-61, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17206408

RESUMEN

Mutations that affect the splicing of pre-mRNA are a major cause of human disease. Familial dysautonomia (FD) is a recessive neurodegenerative disease caused by a T to C transition at base pair 6 of IKBKAP intron 20. This mutation results in variable tissue-specific skipping of exon 20. Previously, we reported that the plant cytokinin kinetin dramatically increases exon 20 inclusion in RNA isolated from cultured FD cells. The goal of the current study was to investigate the nature of the FD splicing defect and the mechanism by which kinetin improves exon inclusion, as such knowledge will facilitate the development of future therapeutics aimed at regulating mRNA splicing. In this study, we demonstrate that treatment of FD lymphoblast cell lines with kinetin increases IKBKAP mRNA and IKAP protein to normal levels. Using a series of minigene constructs, we show that deletion of a region at the end of IKBKAP exon 20 disrupts the ability of kinetin to improve exon inclusion, pinpointing a kinetin responsive sequence element. We next performed a screen of endogenously expressed genes with multiple isoforms resulting from exon skipping events and show that kinetin's ability to improve exon inclusion is not limited to IKBKAP. Lastly, we highlight the potential of kinetin for the treatment of other human splicing disorders by showing correction of a splicing defect in neurofibromatosis.


Asunto(s)
Proteínas Portadoras/genética , Disautonomía Familiar/tratamiento farmacológico , Cinetina/uso terapéutico , Empalme del ARN/efectos de los fármacos , Proteínas Portadoras/análisis , Proteínas Portadoras/efectos de los fármacos , Línea Celular Tumoral , Exones/efectos de los fármacos , Humanos , Cinetina/farmacología , Neurofibromatosis/tratamiento farmacológico , Neurofibromatosis/genética , ARN Mensajero/análisis , ARN Mensajero/efectos de los fármacos , Factores de Elongación Transcripcional
9.
Circulation ; 112(13): 2022-30, 2005 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-16172273

RESUMEN

BACKGROUND: Mitral valve prolapse (MVP) is a common disorder associated with mitral regurgitation, endocarditis, heart failure, and sudden death. To date, 2 MVP loci have been described, but the defective genes have yet to be discovered. In the present study, we analyzed a large family segregating MVP, and identified a new locus, MMVP3. This study and others have enabled us to explore mitral valve morphological variations of currently uncertain clinical significance. METHODS AND RESULTS: Echocardiograms and blood samples were obtained from 43 individuals who were classified by the extent and pattern of displacement. Genotypic analyses were performed with polymorphic microsatellite markers. Evidence of linkage was obtained on chromosome 13q31.3-q32.1, with a peak nonparametric linkage score of 18.41 (P<0.0007). Multipoint parametric analysis gave a logarithm of odds score of 3.17 at marker D13S132. Of the 6 related individuals with mitral valve morphologies not meeting diagnostic criteria but resembling fully developed forms, 5 carried all or part of the haplotype linked to MVP. CONCLUSIONS: The mapping of a new MVP locus to chromosome 13 confirms the observed genetic heterogeneity and represents an important step toward gene identification. Furthermore, the genetic analysis provides clinical lessons with regard to previously nondiagnostic morphologies. In the familial context, these may represent early expression in gene carriers. Early recognition of gene carriers could potentially enhance the clinical evaluation of patients at risk of full expression, with the ultimate aim of developing interventions to reduce progression.


Asunto(s)
Mapeo Cromosómico , Cromosomas Humanos Par 13 , Genes Dominantes , Prolapso de la Válvula Mitral/genética , Adulto , Anciano , Ecocardiografía , Femenino , Heterogeneidad Genética , Ligamiento Genético , Humanos , Escala de Lod , Masculino , Persona de Mediana Edad , Prolapso de la Válvula Mitral/diagnóstico por imagen , Linaje
10.
Brain Res ; 983(1-2): 209-14, 2003 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-12914982

RESUMEN

Familial dysautonomia (FD) is the best-known and most common member of a group of congenital sensory/autonomic neuropathies characterized by widespread sensory and variable autonomic dysfunction. As opposed to the sensory/motor neuropathies, little is known about the causes of neuronal dysfunction and loss in the sensory/autonomic neuropathies. FD involves progressive neuronal degeneration, has a broad impact on the operation of many of the body's systems, and leads to a markedly reduced quality of life and premature death. In 2001, we identified two mutations in the IKBKAP gene that result in FD. IKBKAP encodes IKAP, a member of the putative human holo-Elongator complex, which may facilitate transcription by RNA polymerase II. Whether or not the Elongator plays this role is moot. The FD mutation found on >99.5% of FD chromosomes does not cause complete loss of function. Instead, it results in a tissue-specific decrease in splicing efficiency of the IKBKAP transcript; cells from patients retain some capacity to produce normal mRNA and protein. To better understand the relationship between the genotype of FD patients and their phenotype, we have used in situ hybridization histochemistry to map the IKAP mRNA in sections of whole rat embryos. The mRNA is widely distributed. Highest levels are in the nervous system, but substantial amounts are also present in peripheral organs.


Asunto(s)
Proteínas Portadoras/genética , Disautonomía Familiar/genética , ARN Mensajero/genética , Animales , Autorradiografía , Proteínas Portadoras/biosíntesis , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario y Fetal/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Humanos , Embarazo , ARN Mensajero/biosíntesis , Proteínas de Unión al ARN , Ratas , Distribución Tisular , Factores de Elongación Transcripcional
11.
Mol Cell Biol ; 29(3): 736-44, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19015235

RESUMEN

Familial dysautonomia (FD), a devastating hereditary sensory and autonomic neuropathy, results from an intronic mutation in the IKBKAP gene that disrupts normal mRNA splicing and leads to tissue-specific reduction of IKBKAP protein (IKAP) in the nervous system. To better understand the roles of IKAP in vivo, an Ikbkap knockout mouse model was created. Results from our study show that ablating Ikbkap leads to embryonic lethality, with no homozygous Ikbkap knockout (Ikbkap(-)(/)(-)) embryos surviving beyond 12.5 days postcoitum. Morphological analyses of the Ikbkap(-)(/)(-) conceptus at different stages revealed abnormalities in both the visceral yolk sac and the embryo, including stunted extraembryonic blood vessel formation, delayed entry into midgastrulation, disoriented dorsal primitive neural alignment, and failure to establish the embryonic vascular system. Further, we demonstrate downregulation of several genes that are important for neurulation and vascular development in the Ikbkap(-)(/)(-) embryos and show that this correlates with a defect in transcriptional elongation-coupled histone acetylation. Finally, we show that the embryonic lethality resulting from Ikbkap ablation can be rescued by a human IKBKAP transgene. For the first time, we demonstrate that IKAP is crucial for both vascular and neural development during embryogenesis and that protein function is conserved between mouse and human.


Asunto(s)
Proteínas Portadoras/metabolismo , Pérdida del Embrión/genética , Eliminación de Gen , Subunidades de Proteína/deficiencia , Transcripción Genética , Animales , Vasos Sanguíneos/anomalías , Vasos Sanguíneos/embriología , Cruzamientos Genéticos , Pérdida del Embrión/patología , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Membranas Extraembrionarias/anomalías , Membranas Extraembrionarias/embriología , Femenino , Regulación del Desarrollo de la Expresión Génica , Marcación de Gen , Heterocigoto , Humanos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones , Ratones Noqueados , Subunidades de Proteína/metabolismo , Factores de Elongación Transcripcional , Transgenes
12.
Pediatr Res ; 63(2): 186-90, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18091349

RESUMEN

The common familial dysautonomia (FD) mutation results in tissue specific mis-splicing with reduced amount of wild-type (WT) IkappaB kinase associated protein gene (IKBKAP) mRNA and ELP1. ELP1 is a subunit of Elongator, formerly called the IkappaB kinase associated protein (IKAP) protein. We measured IKBKAP mRNA in peripheral blood leukocytes to determine whether FD subjects and carriers have characteristic levels. Estimated mean IKBKAP mRNA levels, measured by quantitative PCR and expressed as amount relative to the noncarrier average, were significantly different for the two groups when not adjusted for age and sex (p < 0.001): FD subjects 0.23, 95% confidence interval (CI) (0.19, 0.28); carriers 0.58, 95% CI (0.50, 0.68); or adjusted for age and sex (p < 0.001): FD subjects 0.21, 95% CI (0.16, 0.26); carriers 0.66, 95% CI (0.55, 0.79). Comparison of IKBKAP mRNA levels of the 22 FD subjects and their related carriers showed a strong correlation, providing evidence for genetic control of splicing efficiency. IKBKAP mRNA levels were not higher in those subjects using tocotrienols or epigallocatechin gallate. Levels of IKBKAP mRNA in peripheral blood leukocytes can be used to assess molecular response to therapies aimed at enhancing exon 20 inclusion and increasing cellular levels of ELP1/IKAP.


Asunto(s)
Proteínas Portadoras/genética , Disautonomía Familiar/genética , Regulación de la Expresión Génica , Leucocitos/citología , Adolescente , Adulto , Niño , Femenino , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Modelos Biológicos , Modelos Estadísticos , Mutación , Factores de Elongación Transcripcional
13.
Genomics ; 90(3): 389-96, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17644305

RESUMEN

Familial dysautonomia (FD) is a severe hereditary sensory and autonomic neuropathy, and all patients with FD have a splice mutation in the IKBKAP gene. The FD splice mutation results in variable, tissue-specific skipping of exon 20 in IKBKAP mRNA, which leads to reduced IKAP protein levels. The development of therapies for FD will require suitable mouse models for preclinical studies. In this study, we report the generation and characterization of a mouse model carrying the complete human IKBKAP locus with the FD IVS20+6T-->C splice mutation. We show that the mutant IKBKAP transgene is misspliced in this model in a tissue-specific manner that replicates the pattern seen in FD patient tissues. Creation of this humanized mouse is the first step toward development of a complex phenotypic model of FD. These transgenic mice are an ideal model system for testing the effectiveness of therapeutic agents that target the missplicing defect. Last, these mice will permit direct studies of tissue-specific splicing and the identification of regulatory factors that play a role in complex gene expression.


Asunto(s)
Proteínas Portadoras/genética , Mutación , Animales , Disautonomía Familiar/genética , Perfilación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Transgénicos , Modelos Genéticos , Fenotipo , Empalme del ARN , ARN Mensajero/metabolismo , Recombinación Genética , Distribución Tisular , Factores de Elongación Transcripcional
14.
Am J Hum Genet ; 72(3): 749-58, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12577200

RESUMEN

We recently identified a mutation in the I-kappa B kinase associated protein (IKBKAP) gene as the major cause of familial dysautonomia (FD), a recessive sensory and autonomic neuropathy. This alteration, located at base pair 6 of the intron 20 donor splice site, is present on >99.5% of FD chromosomes and results in tissue-specific skipping of exon 20. A second FD mutation, a missense change in exon 19 (R696P), was seen in only four patients heterozygous for the major mutation. Here, we have further characterized the consequences of the major mutation by examining the ratio of wild-type to mutant (WT:MU) IKBKAP transcript in EBV-transformed lymphoblast lines, primary fibroblasts, freshly collected blood samples, and postmortem tissues from patients with FD. We consistently found that WT IKBKAP transcripts were present, albeit to varying extents, in all cell lines, blood, and postmortem FD tissues. Further, a corresponding decrease in the level of WT protein is seen in FD cell lines and tissues. The WT:MU ratio in cultured lymphoblasts varied with growth phase but not with serum concentration or inclusion of antibiotics. Using both densitometry and real-time quantitative polymerase chain reaction, we found that relative WT:MU IKBKAP RNA levels were highest in cultured patient lymphoblasts and lowest in postmortem central and peripheral nervous tissues. These observations suggest that the relative inefficiency of WT IKBKAP mRNA production from the mutant alleles in the nervous system underlies the selective degeneration of sensory and autonomic neurons in FD.Therefore, exploration of methods to increase the WT:MU IKBKAP transcript ratio in the nervous system offers a promising approach for developing an effective therapy for patients with FD.


Asunto(s)
Empalme Alternativo , Proteínas Portadoras/genética , Disautonomía Familiar/genética , Mutación , Línea Celular , Exones , Genes Recesivos , Herpesvirus Humano 4 , Humanos , Linfocitos , Especificidad de Órganos , Transcripción Genética , Factores de Elongación Transcripcional , Transfección
15.
Hum Mol Genet ; 13(4): 429-36, 2004 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-14709595

RESUMEN

The defective splicing of pre-mRNA is a major cause of human disease. Exon skipping is a common result of splice mutations and has been reported in a wide variety of genetic disorders, yet the underlying mechanism is poorly understood. Often, such mutations are incompletely penetrant, and low levels of normal transcript and protein are maintained. Familial dysautonomia (FD) is caused by mutations in IKBKAP, and all cases described to date involve an intron 20 mutation that results in a unique pattern of tissue-specific exon skipping. Accurate splicing of the mutant IKBKAP allele is particularly inefficient in the nervous system. Here we show that treatment with the plant cytokinin kinetin alters splicing of IKBKAP. Kinetin significantly increases inclusion of exon 20 from the endogenous gene, as well as from an IKBKAP minigene. By contrast the drug does not enhance inclusion of alternatively spliced exon 31 in MYO5A. Benzyladenine, the most closely related cytokinin, showed a similar but less dramatic effect. Our findings reveal a remarkable impact on splicing fidelity by these small molecules, which therefore provide new tools for the dissection of mechanisms controlling tissue-specific pre-mRNA splicing. Further, kinetin should be explored as a treatment for increasing the level of normal IKAP in FD, and for other splicing disorders that may share a similar mechanism.


Asunto(s)
Adenina/análogos & derivados , Adenina/farmacología , Empalme Alternativo/efectos de los fármacos , Citocininas/farmacología , Disautonomía Familiar/genética , Precursores del ARN/genética , Empalme Alternativo/genética , Proteínas Portadoras/genética , Células Cultivadas , Exones/genética , Humanos , Cinetina , Cadenas Pesadas de Miosina/genética , Miosina Tipo V/genética , ARN Mensajero/genética , Factores de Elongación Transcripcional
16.
Am J Med Genet A ; 118A(4): 305-8, 2003 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-12687659

RESUMEN

Familial Dysautonomia is an autosomal recessive disease with a remarkably high carrier frequency in the Ashkenazi Jewish population. It has recently been estimated that as many as 1 in 27 Ashkenazi Jews is a carrier of FD. The FD gene has been identified as IKBKAP, and two disease-causing mutations have been identified. The most common mutation, which is present on 99.5% of all FD chromosomes, is an intronic splice site mutation that results in tissue-specific skipping of exon 20. The second mutation, R696P, is a missense mutation that has been identified in 4 unrelated patients heterozygous for the major splice mutation. Interestingly, despite the fact that FD is a recessive disease, normal mRNA and protein are expressed in patient cells. To date, the diagnosis of FD has been limited to individuals of Ashkenazi Jewish descent and identification of the gene has led to widespread diagnostic and carrier testing in this population. In this report, we describe the first non-Jewish IKBKAP mutation, a proline to leucine missense mutation in exon 26, P914L. This mutation is of particular significance because it was identified in a patient who lacks one of the cardinal diagnostic criteria for the disease-pure Ashkenazi Jewish ancestry. In light of this fact, the diagnostic criteria for FD must be expanded. Furthermore, in order to ensure carrier identification in all ethnicities, this mutation must now be considered when screening for FD.


Asunto(s)
Disautonomía Familiar/etnología , Disautonomía Familiar/genética , Mutación Missense/genética , Preescolar , Análisis Mutacional de ADN , Humanos , Judíos/genética , Mapeo Restrictivo
17.
Am J Hum Genet ; 72(6): 1551-9, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12707861

RESUMEN

Mitral valve prolapse (MVP) is a common cardiovascular abnormality in the United States, occurring in approximately 2.4% of the general population. Clinically, patients with MVP exhibit fibromyxomatous changes in one or both of the mitral leaflets that result in superior displacement of the leaflets into the left atrium. Although often clinically benign, MVP can be associated with important accompanying sequelae, including mitral regurgitation, bacterial endocarditis, congestive heart failure, atrial fibrillation, and even sudden death. MVP is genetically heterogeneous and is inherited as an autosomal dominant trait that exhibits both sex- and age-dependent penetrance. In this report, we describe the results of a genome scan and show that a locus for MVP maps to chromosome 11p15.4. Multipoint parametric analysis performed by use of GENEHUNTER gave a maximum LOD score of 3.12 for the chromosomal region immediately surrounding the four-marker haplotype D11S4124-D11S2349-D11S1338-D11S1323, and multipoint nonparametric analysis (NPL) confirms this finding (NPL=38.59; P=.000397). Haplotype analysis across this region defines a 4.3-cM region between the markers D11S1923 and D11S1331 as the location of a new MVP locus, MMVP2, and confirms the genetic heterogeneity of this disorder. The discovery of genes involved in the pathogenesis of this common disease is crucial to understanding the marked variability in disease expression and mortality seen in MVP.


Asunto(s)
Cromosomas Humanos Par 11 , Genes Dominantes , Prolapso de la Válvula Mitral/genética , Adolescente , Adulto , Anciano , Niño , Preescolar , Femenino , Efecto Fundador , Heterogeneidad Genética , Ligamiento Genético , Marcadores Genéticos , Pruebas Genéticas/métodos , Haplotipos , Humanos , Masculino , Persona de Mediana Edad , Linaje
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA