Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Methods Mol Biol ; 1470: 151-62, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27581291

RESUMEN

RNA interference (RNAi) is a process in which double-stranded RNA (dsRNA) molecules mediate the inhibition of gene expression. RNAi in C. elegans can be achieved by simply feeding animals with bacteria expressing dsRNA against the gene of interest. This "feeding" method has made it possible to conduct genome-wide RNAi experiments for the systematic knockdown and subsequent investigation of almost every single gene in the genome. Historically, these genome-scale RNAi screens have been labor and time intensive. However, recent advances in automated, high-throughput methodologies have allowed the development of more rapid and efficient screening protocols. In this report, we describe a fast and efficient, liquid-based method for genome-wide RNAi screening.


Asunto(s)
Caenorhabditis elegans/genética , Ensayos Analíticos de Alto Rendimiento/métodos , Interferencia de ARN , Animales , Genoma de los Helmintos , Procesamiento de Imagen Asistido por Computador
2.
PLoS One ; 10(10): e0141542, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26512890

RESUMEN

α1-antitrypsin deficiency (ATD) predisposes patients to both loss-of-function (emphysema) and gain-of-function (liver cirrhosis) phenotypes depending on the type of mutation. Although the Z mutation (ATZ) is the most prevalent cause of ATD, >120 mutant alleles have been identified. In general, these mutations are classified as deficient (<20% normal plasma levels) or null (<1% normal levels) alleles. The deficient alleles, like ATZ, misfold in the ER where they accumulate as toxic monomers, oligomers and aggregates. Thus, deficient alleles may predispose to both gain- and loss-of-function phenotypes. Null variants, if translated, typically yield truncated proteins that are efficiently degraded after being transiently retained in the ER. Clinically, null alleles are only associated with the loss-of-function phenotype. We recently developed a C. elegans model of ATD in order to further elucidate the mechanisms of proteotoxicity (gain-of-function phenotype) induced by the aggregation-prone deficient allele, ATZ. The goal of this study was to use this C. elegans model to determine whether different types of deficient and null alleles, which differentially affect polymerization and secretion rates, correlated to any extent with proteotoxicity. Animals expressing the deficient alleles, Mmalton, Siiyama and S (ATS), showed overall toxicity comparable to that observed in patients. Interestingly, Siiyama expressing animals had smaller intracellular inclusions than ATZ yet appeared to have a greater negative effect on animal fitness. Surprisingly, the null mutants, although efficiently degraded, showed a relatively mild gain-of-function proteotoxic phenotype. However, since null variant proteins are degraded differently and do not appear to accumulate, their mechanism of proteotoxicity is likely to be different to that of polymerizing, deficient mutants. Taken together, these studies showed that C. elegans is an inexpensive tool to assess the proteotoxicity of different AT variants using a transgenic approach.


Asunto(s)
Caenorhabditis elegans/metabolismo , Mutación , Serpinas/genética , Deficiencia de alfa 1-Antitripsina/genética , Alelos , Secuencia de Aminoácidos , Animales , Caenorhabditis elegans/genética , Datos de Secuencia Molecular , Transporte de Proteínas , Proteolisis , Serpinas/metabolismo , Serpinas/toxicidad , Deficiencia de alfa 1-Antitripsina/metabolismo
3.
Curr Protoc Cytom ; 74: 12.41.1-12.41.17, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26423692

RESUMEN

Caenorhabditis elegans is a powerful model organism for studying human biology and disease due to its surprisingly high genetic homology to Homo sapiens. Its genetic amenability, small size, short generation time, and transparent body make it an ideal organism for multiple scientific disciplines. Fluorescent microscopy is essential for studying protein biological function. However, C. elegans, mainly due to its high motility, has been more difficult to adapt to fluorescence imaging, especially live-imaging. We present here several protocols for the study of protein location, function and dynamics in context of a whole animal. These protocols, especially when combined with existing genetic procedures, can yield a great deal of insight in the physiological roles of proteins in C. elegans, which can be directly translated into mammalian systems.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Animales , Caenorhabditis elegans/citología , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Humanos , Microscopía Fluorescente/métodos , Transporte de Proteínas/fisiología
4.
Genetics ; 200(1): 207-19, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25786854

RESUMEN

Familial encephalopathy with neuroserpin inclusions bodies (FENIB) is a serpinopathy that induces a rare form of presenile dementia. Neuroserpin contains a classical signal peptide and like all extracellular serine proteinase inhibitors (serpins) is secreted via the endoplasmic reticulum (ER)-Golgi pathway. The disease phenotype is due to gain-of-function missense mutations that cause neuroserpin to misfold and aggregate within the ER. In a previous study, nematodes expressing a homologous mutation in the endogenous Caenorhabditis elegans serpin, srp-2, were reported to model the ER proteotoxicity induced by an allele of mutant neuroserpin. Our results suggest that SRP-2 lacks a classical N-terminal signal peptide and is a member of the intracellular serpin family. Using confocal imaging and an ER colocalization marker, we confirmed that GFP-tagged wild-type SRP-2 localized to the cytosol and not the ER. Similarly, the aggregation-prone SRP-2 mutant formed intracellular inclusions that localized to the cytosol. Interestingly, wild-type SRP-2, targeted to the ER by fusion to a cleavable N-terminal signal peptide, failed to be secreted and accumulated within the ER lumen. This ER retention phenotype is typical of other obligate intracellular serpins forced to translocate across the ER membrane. Neuroserpin is a secreted protein that inhibits trypsin-like proteinase. SRP-2 is a cytosolic serpin that inhibits lysosomal cysteine peptidases. We concluded that SRP-2 is neither an ortholog nor a functional homolog of neuroserpin. Furthermore, animals expressing an aggregation-prone mutation in SRP-2 do not model the ER proteotoxicity associated with FENIB.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Retículo Endoplásmico/metabolismo , Serpinas/metabolismo , Secuencia de Aminoácidos , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/genética , Citosol/metabolismo , Datos de Secuencia Molecular , Agregado de Proteínas/genética , Señales de Clasificación de Proteína , Transporte de Proteínas , Serpinas/química , Serpinas/genética
5.
Methods ; 68(3): 529-35, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24990146

RESUMEN

Recent breakthrough discoveries have shown that committed cell fates can be reprogrammed by genetic, chemical and environmental manipulations. The germline of the nematode Caenorhabditis elegans provides a tractable system for studying cell fate reprogramming within the context of a whole organism. To explore the possibility of using C. elegans in high-throughput screens (HTS), we developed a high-throughput workflow for testing compounds that modulate cell fate reprogramming. We utilized puf-8; lip-1 mutants that have enhanced MPK-1 (an ERK homolog)/MAP kinase (MAPK) signaling. Wild-type C. elegans hermaphrodites produce both sperm and oocytes, and are thus self-fertile. However, puf-8; lip-1 mutants produce only sperm and are sterile. Notably, compounds that pharmacologically down-regulate MPK-1 (an ERK homolog)/MAP kinase (MAPK) signaling are able to reprogram germ cell fate and restore fertility to these animals. puf-8; lip-1 mutants provide numerous challenges for HTS. First, they are sterile as homozygotes and must be maintained as heterozygotes using a balancer chromosome. Second, homozygous animals for experimentation must be physically separated from the rest of the population. Third, a high quality, high-content assay has not been developed to measure compound effects on germ cell fate reprogramming. Here we describe a semi-automated high-throughput workflow that enables effective sorting of homozygous puf-8; lip-1 mutants into 384-well plates using the COPAS™ BIOSORT. In addition, we have developed an image-based assay for rapidly measuring germ cell reprogramming by measuring the number of viable progeny in wells. The methods presented in this report enable the use of puf-8; lip-1 mutants in HTS campaigns for chemical modulators of germ cell reprogramming within the context of a whole organism.


Asunto(s)
Caenorhabditis elegans/efectos de los fármacos , Reprogramación Celular/efectos de los fármacos , Células Germinativas/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Animales , Caenorhabditis elegans/crecimiento & desarrollo , Linaje de la Célula/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento , Masculino , Transducción de Señal/efectos de los fármacos
6.
Expert Opin Drug Discov ; 9(9): 1021-32, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24998976

RESUMEN

INTRODUCTION: Many human diseases result from a failure of a single protein to achieve the correct folding and tertiary conformation. These so-called 'conformational diseases' involve diverse proteins and distinctive cellular pathologies. They all engage the proteostasis network (PN), to varying degrees in an attempt to mange cellular stress and restore protein homeostasis. The insulin/insulin-like growth factor signaling (IIS) pathway is a master regulator of cellular stress response, which is implicated in regulating components of the PN. AREAS COVERED: This review focuses on novel approaches to target conformational diseases. The authors discuss the evidence supporting the involvement of the IIS pathway in modulating the PN and regulating proteostasis in Caenorhabditis elegans. Furthermore, they review previous PN and IIS drug screens and explore the possibility of using C. elegans for whole organism-based drug discovery for modulators of IIS-proteostasis pathways. EXPERT OPINION: An alternative approach to develop individualized therapy for each conformational disease is to modulate the global PN. The involvement of the IIS pathway in regulating longevity and response to a variety of stresses is well documented. Increasing data now provide evidence for the close association between the IIS and the PN pathways. The authors believe that high-throughput screening campaigns, which target the C. elegans IIS pathway, may identify drugs that are efficacious in treating numerous conformational diseases.


Asunto(s)
Caenorhabditis elegans/fisiología , Descubrimiento de Drogas/métodos , Estrés Fisiológico/fisiología , Animales , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Insulina/fisiología , Longevidad/fisiología , Pliegue de Proteína , Estructura Terciaria de Proteína/fisiología , Transducción de Señal/fisiología , Somatomedinas/fisiología
7.
Hum Mol Genet ; 23(19): 5109-22, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24838286

RESUMEN

The accumulation of serpin oligomers and polymers within the endoplasmic reticulum (ER) causes cellular injury in patients with the classical form α1-antitrypsin deficiency (ATD). To better understand the cellular and molecular genetic aspects of this disorder, we generated transgenic C. elegans strains expressing either the wild-type (ATM) or Z mutant form (ATZ) of the human serpin fused to GFP. Animals secreted ATM, but retained polymerized ATZ within dilated ER cisternae. These latter animals also showed slow growth, smaller brood sizes and decreased longevity; phenotypes observed in ATD patients or transgenic mouse lines expressing ATZ. Similar to mammalian models, ATZ was disposed of by autophagy and ER-associated degradation pathways. Mutant strains defective in insulin signaling (daf-2) also showed a marked decrease in ATZ accumulation. Enhanced ATZ turnover was associated with the activity of two proteins central to systemic/exogenous (exo)-RNAi pathway: the dsRNA importer, SID-1 and the argonaute, RDE-1. Animals with enhanced exo-RNAi activity (rrf-3 mutant) phenocopied the insulin signaling mutants and also showed increased ATZ turnover. Taken together, these studies allude to the existence of a novel proteostasis pathway that mechanistically links misfolded protein turnover to components of the systemic RNAi machinery.


Asunto(s)
Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Interferencia de ARN , Deficiencia de alfa 1-Antitripsina/genética , alfa 1-Antitripsina/genética , Animales , Animales Modificados Genéticamente , Proteínas de Caenorhabditis elegans/genética , Línea Celular , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/ultraestructura , Degradación Asociada con el Retículo Endoplásmico , Expresión Génica , Genes Reporteros , Humanos , Insulina/metabolismo , Ratones , Ratones Transgénicos , Mutación , Fenotipo , Regiones Promotoras Genéticas , Proteolisis , Deficiencias en la Proteostasis/genética , Deficiencias en la Proteostasis/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Serpinas , Transducción de Señal , Intercambiadores de Sodio-Hidrógeno/genética , alfa 1-Antitripsina/metabolismo , Deficiencia de alfa 1-Antitripsina/metabolismo
8.
Hum Mol Genet ; 23(19): 5123-32, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24838285

RESUMEN

α1-Antitrypsin deficiency (ATD) is a common genetic disorder that can lead to end-stage liver and lung disease. Although liver transplantation remains the only therapy currently available, manipulation of the proteostasis network (PN) by small molecule therapeutics offers great promise. To accelerate the drug-discovery process for this disease, we first developed a semi-automated high-throughput/content-genome-wide RNAi screen to identify PN modifiers affecting the accumulation of the α1-antitrypsin Z mutant (ATZ) in a Caenorhabditis elegans model of ATD. We identified 104 PN modifiers, and these genes were used in a computational strategy to identify human ortholog-ligand pairs. Based on rigorous selection criteria, we identified four FDA-approved drugs directed against four different PN targets that decreased the accumulation of ATZ in C. elegans. We also tested one of the compounds in a mammalian cell line with similar results. This methodology also proved useful in confirming drug targets in vivo, and predicting the success of combination therapy. We propose that small animal models of genetic disorders combined with genome-wide RNAi screening and computational methods can be used to rapidly, economically and strategically prime the preclinical discovery pipeline for rare and neglected diseases with limited therapeutic options.


Asunto(s)
Descubrimiento de Drogas , Estudio de Asociación del Genoma Completo , Interferencia de ARN , Deficiencia de alfa 1-Antitripsina/genética , alfa 1-Antitripsina/genética , Animales , Caenorhabditis elegans , Biología Computacional , Modelos Animales de Enfermedad , Genómica , Ensayos Analíticos de Alto Rendimiento , Humanos , Mutación , Unión Proteica , Deficiencias en la Proteostasis/genética , Reproducibilidad de los Resultados , Deficiencia de alfa 1-Antitripsina/tratamiento farmacológico
9.
Methods ; 68(3): 542-7, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24836996

RESUMEN

There are many challenges to live Caenorhabditis elegans imaging including the high motility of the animals and sustaining their viability for extended periods of time. Commonly used anesthetics to immobilize the C. elegans for imaging purpose prevents feeding of the animals and can cause cellular physiologic changes. Here we present three adapted or novel methodologies to image live C. elegans over different imaging microscopy equipment to allow for visualization of animals by DIC and fluorescence without the use of microfluidic technologies. The methods present here use common microscopy consumables and equipment found in many imaging core facilities and can be easily adapted to fit on multiple microscopy systems.


Asunto(s)
Microscopía Fluorescente/métodos , Imagen Óptica/métodos , Animales , Caenorhabditis elegans
10.
PLoS One ; 9(1): e87260, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24498058

RESUMEN

The classical form of α1-antitrypsin deficiency (ATD) is associated with hepatic fibrosis and hepatocellular carcinoma. It is caused by the proteotoxic effect of a mutant secretory protein that aberrantly accumulates in the endoplasmic reticulum of liver cells. Recently we developed a model of this deficiency in C. elegans and adapted it for high-content drug screening using an automated, image-based array scanning. Screening of the Library of Pharmacologically Active Compounds identified fluphenazine (Flu) among several other compounds as a drug which reduced intracellular accumulation of mutant α1-antitrypsin Z (ATZ). Because it is representative of the phenothiazine drug class that appears to have autophagy enhancer properties in addition to mood stabilizing activity, and can be relatively easily re-purposed, we further investigated its effects on mutant ATZ. The results indicate that Flu reverses the phenotypic effects of ATZ accumulation in the C. elegans model of ATD at doses which increase the number of autophagosomes in vivo. Furthermore, in nanomolar concentrations, Flu enhances the rate of intracellular degradation of ATZ and reduces the cellular ATZ load in mammalian cell line models. In the PiZ mouse model Flu reduces the accumulation of ATZ in the liver and mediates a decrease in hepatic fibrosis. These results show that Flu can reduce the proteotoxicity of ATZ accumulation in vivo and, because it has been used safely in humans, this drug can be moved rapidly into trials for liver disease due to ATD. The results also provide further validation for drug discovery using C. elegans models that can be adapted to high-content drug screening platforms and used together with mammalian cell line and animal models.


Asunto(s)
Caenorhabditis elegans/metabolismo , Modelos Animales de Enfermedad , Flufenazina/farmacología , Deficiencia de alfa 1-Antitripsina/prevención & control , Animales , Animales Modificados Genéticamente , Antipsicóticos/farmacología , Autofagia/efectos de los fármacos , Autofagia/genética , Células CHO , Caenorhabditis elegans/genética , Cricetinae , Cricetulus , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Células Hep G2 , Humanos , Immunoblotting , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Mutación , Fagosomas/efectos de los fármacos , Fagosomas/metabolismo , Análisis de Supervivencia , alfa 1-Antitripsina/genética , alfa 1-Antitripsina/metabolismo , Deficiencia de alfa 1-Antitripsina/genética , Deficiencia de alfa 1-Antitripsina/metabolismo
11.
Adv Drug Deliv Rev ; 69-70: 247-53, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24333896

RESUMEN

Caenorhabditis elegans has been proven to be a useful model organism for investigating molecular and cellular aspects of numerous human diseases. More recently, investigators have explored the use of this organism as a tool for drug discovery. Although earlier drug screens were labor-intensive and low in throughput, recent advances in high-throughput liquid workflows, imaging platforms and data analysis software have made C. elegans a viable option for automated high-throughput drug screens. This review will outline the evolution of C. elegans-based drug screening, discuss the inherent challenges of using C. elegans, and highlight recent technological advances that have paved the way for future drug screens.


Asunto(s)
Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/fisiología , Descubrimiento de Drogas/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Animales , Descubrimiento de Drogas/tendencias , Ensayos Analíticos de Alto Rendimiento/tendencias , Humanos
12.
Int J Biochem Cell Biol ; 47: 109-12, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24355812

RESUMEN

Hepatocytes are metabolically active cells of the liver that play an important role in the biosynthesis of proteins including α1-antitrypsin. Mutations in the α1-antitrypsin gene can lead to protein misfolding, polymerization/aggregation and retention of protein within the endoplasmic reticulum of hepatocytes. The intracellular accumulation of α1-antitrypsin aggregates can lead to liver disease and increased likelihood of developing hepatocellular carcinomas. Of note, only ~10% of individuals with α1-antitrypsin-deficiency develop severe liver disease suggesting that there are other genetic and/or environmental factors that determine disease outcome. The nematode, Caenorhabditis elegans, is a powerful genetic model organism to study molecular aspects of human disease. In this review, we discuss the functional similarities between the intestinal cells of C. elegans and human hepatocytes and how a C. elegans model of α1-antitrypsin-deficiency can be used as a tool for identifying genetic modifiers and small molecule drugs.


Asunto(s)
Caenorhabditis elegans , Modelos Animales de Enfermedad , Hepatocitos/patología , Deficiencia de alfa 1-Antitripsina/tratamiento farmacológico , Deficiencia de alfa 1-Antitripsina/patología , Animales , Descubrimiento de Drogas , Hepatocitos/metabolismo , Humanos , Deficiencia de alfa 1-Antitripsina/metabolismo
13.
Cell Signal ; 25(4): 860-6, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23266470

RESUMEN

Timeless was originally identified in Drosophila as an essential component of circadian cycle regulation, where its function is tightly controlled at the protein level by tyrosine phosphorylation and subsequent degradation. In mammals, Timeless has also been implicated in circadian rhythms as well as cell cycle control and embryonic development. Here we report that mammalian Timeless is an SH3 domain-binding protein and substrate for several members of the Src protein-tyrosine kinase family, including Fyn, Hck, c-Src and c-Yes. Co-expression of Tim with Fyn or Hck was followed by ubiquitylation and subsequent degradation in human 293T cells. While c-Src and c-Yes also promoted Tim ubiquitylation, in this case ubiquitylation correlated with Tim protein accumulation rather than degradation. Both c-Src and c-Yes selectively promoted modification of Tim through Lys63-linked polyubiquitin, which may explain the differential effects on Tim protein turnover. These data show distinct and opposing roles for individual Src-family members in the regulation of Tim protein levels, suggesting a unique mechanism for the regulation of Tim function in mammals.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Familia-src Quinasas/metabolismo , Animales , Proteína Tirosina Quinasa CSK , Proteínas de Ciclo Celular/genética , Proteínas de Drosophila , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Proteínas Proto-Oncogénicas c-fyn/química , Proteínas Proto-Oncogénicas c-fyn/genética , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Proteínas Proto-Oncogénicas c-hck/química , Proteínas Proto-Oncogénicas c-hck/genética , Proteínas Proto-Oncogénicas c-hck/metabolismo , Proteínas Proto-Oncogénicas c-yes/química , Proteínas Proto-Oncogénicas c-yes/genética , Proteínas Proto-Oncogénicas c-yes/metabolismo , Especificidad por Sustrato , Transfección , Ubiquitinación , Dominios Homologos src , Familia-src Quinasas/química , Familia-src Quinasas/genética
14.
PLoS One ; 6(2): e17157, 2011 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-21359199

RESUMEN

BACKGROUND: Programmed cell death is critical not only in adult tissue homeostasis but for embryogenesis as well. One of the earliest steps in development, formation of the proamniotic cavity, involves coordinated apoptosis of embryonic cells. Recent work from our group demonstrated that c-Src protein-tyrosine kinase activity triggers differentiation of mouse embryonic stem (mES) cells to primitive ectoderm-like cells. In this report, we identified Timeless (Tim), the mammalian ortholog of a Drosophila circadian rhythm protein, as a binding partner and substrate for c-Src and probed its role in the differentiation of mES cells. METHODOLOGY/PRINCIPAL FINDINGS: To determine whether Tim is involved in ES cell differentiation, Tim protein levels were stably suppressed using shRNA. Tim-defective ES cell lines were then tested for embryoid body (EB) formation, which models early mammalian development. Remarkably, confocal microscopy revealed that EBs formed from the Tim-knockdown ES cells failed to cavitate. Cells retained within the centers of the failed cavities strongly expressed the pluripotency marker Oct4, suggesting that further development is arrested without Tim. Immunoblots revealed reduced basal Caspase activity in the Tim-defective EBs compared to wild-type controls. Furthermore, EBs formed from Tim-knockdown cells demonstrated resistance to staurosporine-induced apoptosis, consistent with a link between Tim and programmed cell death during cavitation. CONCLUSIONS/SIGNIFICANCE: Our data demonstrate a novel function for the clock protein Tim during a key stage of early development. Specifically, EBs formed from ES cells lacking Tim showed reduced caspase activity and failed to cavitate. As a consequence, further development was halted, and the cells present in the failed cavity remained pluripotent. These findings reveal a new function for Tim in the coordination of ES cell differentiation, and raise the intriguing possibility that circadian rhythms and early development may be intimately linked.


Asunto(s)
Apoptosis/genética , Tipificación del Cuerpo/genética , Proteínas de Ciclo Celular/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Secuencia de Aminoácidos , Animales , Apoptosis/efectos de los fármacos , Tipificación del Cuerpo/efectos de los fármacos , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Cultivadas , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/fisiología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/fisiología , Técnicas de Inactivación de Genes , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Datos de Secuencia Molecular , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Proteínas Circadianas Period/fisiología , ARN Interferente Pequeño/farmacología
15.
J Mol Biol ; 383(2): 414-23, 2008 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-18775435

RESUMEN

Recent studies have shown that trans-phosphorylation of the Abl SH3 domain at Tyr89 by Src-family kinases is required for the full transforming activity of Bcr-Abl. Tyr89 localizes to a binding surface of the SH3 domain that engages the SH2-kinase linker in the crystal structure of the c-Abl core. Displacement of SH3 from the linker is likely to influence efficient downregulation of c-Abl. Hydrogen-deuterium exchange (HX) and mass spectrometry (MS) were used to investigate whether Tyr89 phosphorylation affects the ability of the SH3 domain to interact intramolecularly with the SH2-kinase linker in cis as well as other peptide ligands in trans. HX MS analysis of SH3 binding showed that when various Abl constructs were phosphorylated at Tyr89 by the Src-family kinase Hck, SH3 was unable to engage a high-affinity ligand in trans and that interaction with the linker in cis was reduced dramatically in a construct containing the SH3 and SH2 domains plus the linker. Phosphorylation of the Abl SH3 domain on Tyr89 also interfered with binding to the negative regulatory protein Abi-1 in trans. Site-directed mutagenesis of Tyr89 and Tyr245, another tyrosine phosphorylation site located in the linker that may also influence SH3 binding, implicated Tyr89 as the key residue necessary for disrupting regulation after phosphorylation. These results imply that phosphorylation at Tyr89 by Src-family kinases prevents engagement of the Abl SH3 domain with its intramolecular binding partner leading to enhanced Abl kinase activity and cellular signaling.


Asunto(s)
Proteínas Proto-Oncogénicas c-abl/química , Proteínas Proto-Oncogénicas c-abl/metabolismo , Tirosina/metabolismo , Dominios Homologos src , Secuencia de Aminoácidos , Sitios de Unión , Proteínas de Fusión bcr-abl/química , Proteínas de Fusión bcr-abl/metabolismo , Genes abl , Humanos , Espectrometría de Masas , Modelos Moleculares , Datos de Secuencia Molecular , Fosforilación , Conformación Proteica , Transducción de Señal , Tirosina/genética , Dominios Homologos src/genética
16.
FEBS J ; 272(17): 4549-57, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16128823

RESUMEN

Two novel rare mutations, MCAD approximately 842G-->C (R256T) and MCAD approximately 1166A-->G (K364R), have been investigated to assess how far the biochemical properties of the mutant proteins correlate with the clinical phenotype of medium chain acyl-CoA dehydrogenase (MCAD) deficiency. When the gene for K364R was overexpressed in Escherichia coli, the synthesized mutant protein only exhibited activity when the gene for chaperonin GroELS was co-overexpressed. Levels of activity correlated with the amounts of native MCAD protein visible in western blots. The R256T mutant, by contrast, displayed no activity either with or without chaperonin, but in this case a strong MCAD protein band was seen in the western blots throughout. The proteins were also purified, and the enzyme function and thermostability investigated. The K364R protein showed only moderate kinetic impairment, whereas the R256T protein was again totally inactive. Neither mutant showed marked depletion of FAD. The pure K364R protein was considerably less thermostable than wild-type MCAD. Western blots indicated that, although the R256T mutant protein is less thermostable than normal MCAD, it is much more stable than K364R. Though clinically asymptomatic thus far, both mutations have a severe impact on the biochemical phenotype of the protein. K364R, like several previously described MCAD mutant proteins, appears to be defective in folding. R256T, by contrast, is a well-folded protein that is nevertheless devoid of catalytic activity. How the mutations specifically affect the catalytic activity and the folding is further discussed.


Asunto(s)
Acil-CoA Deshidrogenasa/química , Acil-CoA Deshidrogenasa/genética , Acil-CoA Deshidrogenasa/deficiencia , Acil-CoA Deshidrogenasa/metabolismo , Sustitución de Aminoácidos , Secuencia de Bases , Dominio Catalítico/genética , Chaperoninas/farmacología , ADN/genética , Estabilidad de Enzimas , Escherichia coli/enzimología , Escherichia coli/genética , Flavina-Adenina Dinucleótido/metabolismo , Humanos , Técnicas In Vitro , Cinética , Modelos Moleculares , Fenotipo , Mutación Puntual , Pliegue de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Espectrofotometría
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...