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










Base de datos
Intervalo de año de publicación
1.
Ann Hum Genet ; 88(1): 86-100, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37921557

RESUMEN

INTRODUCTION: Joubert syndrome (JS) arises from defects of primary cilia resulting in potential malformations of the brain, kidneys, eyes, liver, and limbs. Several of the 35+ genes associated with JS have recognized genotype/phenotype correlations, but most genes have not had enough reported individuals to draw meaningful conclusions. METHODS: A PubMed literature review identified 688 individuals with JS across 32 genes and 112 publications to bolster known genotype/phenotype relationships and identify new correlations. All included patients had the "molar tooth sign" and a confirmed genetic diagnosis. Individuals were categorized by age, ethnicity, sex and the presence of developmental disability/intellectual disability, hypotonia, abnormal eye movements, ataxia, visual impairment, renal impairment, polydactyly, and liver abnormalities. RESULTS: Most genes demonstrated unique phenotypic profiles. Grouping proteins based on physiologic interactions established stronger phenotypic relationships that reflect known ciliary pathophysiology. Age-stratified data demonstrated that end-organ disease is progressive in JS. Most genes demonstrated a significant skew towards having variants with either residual protein function or no residual protein function. CONCLUSION: This cohort demonstrates that clinically meaningful genotype/phenotype relationships exist within most JS-related genes and can be referenced to allow for more personalized clinical care.


Asunto(s)
Anomalías Múltiples , Anomalías del Ojo , Enfermedades Renales Quísticas , Humanos , Anomalías Múltiples/genética , Cerebelo/anomalías , Enfermedades Renales Quísticas/genética , Anomalías del Ojo/genética , Retina/anomalías , Proteínas/genética , Variación Biológica Poblacional
2.
iScience ; 26(7): 106909, 2023 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-37332674

RESUMEN

Characterizing perturbation of molecular pathways in congenital Zika virus (ZIKV) infection is critical for improved therapeutic approaches. Leveraging integrative systems biology, proteomics, and RNA-seq, we analyzed embryonic brain tissues from an immunocompetent, wild-type congenital ZIKV infection mouse model. ZIKV induced a robust immune response accompanied by the downregulation of critical neurodevelopmental gene programs. We identified a negative correlation between ZIKV polyprotein abundance and host cell cycle-inducing proteins. We further captured the downregulation of genes/proteins, many of which are known to be causative for human microcephaly, including Eomesodermin/T-box Brain Protein 2 (EOMES/TBR2) and Neuronal Differentiation 2 (NEUROD2). Disturbances of distinct molecular pathways in neural progenitors and post-mitotic neurons may contribute to complex brain phenotype of congenital ZIKV infection. Overall, this report on protein- and transcript-level dynamics enhances understanding of the ZIKV immunopathological landscape through characterization of fetal immune response in the developing brain.

3.
EMBO Rep ; 23(9): e52211, 2022 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-35793002

RESUMEN

Zika virus (ZIKV) is a flavivirus transmitted via mosquitoes and sex to cause congenital neurodevelopmental defects, including microcephaly. Inherited forms of microcephaly (MCPH) are associated with disrupted centrosome organization. Similarly, we found that ZIKV infection disrupted centrosome organization. ZIKV infection disrupted the organization of centrosomal proteins including CEP63, a MCPH-associated protein. The ZIKV nonstructural protein NS3 bound CEP63, and expression of NS3 was sufficient to alter centrosome architecture and CEP63 localization. Loss of CEP63 suppressed ZIKV-induced centrosome disorganization, indicating that ZIKV requires CEP63 to disrupt centrosome organization. ZIKV infection or CEP63 loss decreased the centrosomal localization and stability of TANK-binding kinase 1 (TBK1), a regulator of the innate immune response. ZIKV infection also increased the centrosomal accumulation of the CEP63 interactor DTX4, a ubiquitin ligase that degrades TBK1. Therefore, we propose that ZIKV disrupts CEP63 function to increase centrosomal DTX4 localization and destabilization of TBK1, thereby tempering the innate immune response.


Asunto(s)
Microcefalia , Infección por el Virus Zika , Virus Zika , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Centrosoma/metabolismo , Humanos , Inmunidad Innata , Microcefalia/metabolismo , Virus Zika/fisiología
4.
Sci Transl Med ; 12(536)2020 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-32213627

RESUMEN

The emergence of CRISPR-Cas9 gene-editing technologies and genome-wide CRISPR-Cas9 libraries enables efficient unbiased genetic screening that can accelerate the process of therapeutic discovery for genetic disorders. Here, we demonstrate the utility of a genome-wide CRISPR-Cas9 loss-of-function library to identify therapeutic targets for facioscapulohumeral muscular dystrophy (FSHD), a genetically complex type of muscular dystrophy for which there is currently no treatment. In FSHD, both genetic and epigenetic changes lead to misexpression of DUX4, the FSHD causal gene that encodes the highly cytotoxic DUX4 protein. We performed a genome-wide CRISPR-Cas9 screen to identify genes whose loss-of-function conferred survival when DUX4 was expressed in muscle cells. Genes emerging from our screen illuminated a pathogenic link to the cellular hypoxia response, which was revealed to be the main driver of DUX4-induced cell death. Application of hypoxia signaling inhibitors resulted in increased DUX4 protein turnover and subsequent reduction of the cellular hypoxia response and cell death. In addition, these compounds proved successful in reducing FSHD disease biomarkers in patient myogenic lines, as well as improving structural and functional properties in two zebrafish models of FSHD. Our genome-wide perturbation of pathways affecting DUX4 expression has provided insight into key drivers of DUX4-induced pathogenesis and has identified existing compounds with potential therapeutic benefit for FSHD. Our experimental approach presents an accelerated paradigm toward mechanistic understanding and therapeutic discovery of a complex genetic disease, which may be translatable to other diseases with well-established phenotypic selection assays.


Asunto(s)
Distrofia Muscular Facioescapulohumeral , Animales , Sistemas CRISPR-Cas/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Distrofia Muscular Facioescapulohumeral/genética , Distrofia Muscular Facioescapulohumeral/terapia , Pez Cebra/genética , Pez Cebra/metabolismo
5.
Neuron ; 106(2): 246-255.e6, 2020 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-32097629

RESUMEN

Genes mutated in human neuronal migration disorders encode tubulin proteins and a variety of tubulin-binding and -regulating proteins, but it is very poorly understood how these proteins function together to coordinate migration. Additionally, the way in which regional differences in neocortical migration are controlled is completely unknown. Here we describe a new syndrome with remarkably region-specific effects on neuronal migration in the posterior cortex, reflecting de novo variants in CEP85L. We show that CEP85L is required cell autonomously in vivo and in vitro for migration, that it localizes to the maternal centriole, and that it forms a complex with many other proteins required for migration, including CDK5, LIS1, NDE1, KIF2A, and DYNC1H1. Loss of CEP85L disrupts CDK5 localization and activation, leading to centrosome disorganization and disrupted microtubule cytoskeleton organization. Together, our findings suggest that CEP85L highlights a complex that controls CDK5 activity to promote neuronal migration.


Asunto(s)
Movimiento Celular , Quinasa 5 Dependiente de la Ciclina/genética , Proteínas del Citoesqueleto/genética , Lisencefalia/genética , Lisencefalia/patología , Neocórtex/patología , Neuronas/patología , Proteínas de Fusión Oncogénica/genética , Centriolos/genética , Niño , Preescolar , Femenino , Humanos , Masculino , Microtúbulos/genética , Microtúbulos/ultraestructura , Proteínas del Tejido Nervioso/fisiología , Adulto Joven
6.
J Cell Biol ; 218(7): 2185-2197, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31197030

RESUMEN

In mammals, centrioles participate in brain development, and human mutations affecting centriole duplication cause microcephaly. Here, we identify a role for the mammalian homologue of yeast SFI1, involved in the duplication of the yeast spindle pole body, as a critical regulator of centriole duplication in mammalian cells. Mammalian SFI1 interacts with USP9X, a deubiquitylase associated with human syndromic mental retardation. SFI1 localizes USP9X to the centrosome during S phase to deubiquitylate STIL, a critical regulator of centriole duplication. USP9X-mediated deubiquitylation protects STIL from degradation. Consistent with a role for USP9X in stabilizing STIL, cells from patients with USP9X loss-of-function mutations have reduced STIL levels. Together, these results demonstrate that SFI1 is a centrosomal protein that localizes USP9X to the centrosome to stabilize STIL and promote centriole duplication. We propose that the USP9X protection of STIL to facilitate centriole duplication underlies roles of both proteins in human neurodevelopment.


Asunto(s)
Proteínas de Ciclo Celular/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Microcefalia/genética , Ubiquitina Tiolesterasa/genética , Ciclo Celular/genética , Centriolos/genética , Centriolos/ultraestructura , Centrosoma/ultraestructura , Femenino , Fibroblastos/metabolismo , Células HEK293 , Células HeLa , Humanos , Microcefalia/patología , Microscopía Electrónica , Mutación , Trastornos del Neurodesarrollo/genética , Proteolisis
7.
Nature ; 556(7701): 370-375, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29643508

RESUMEN

The human cerebral cortex is distinguished by its large size and abundant gyrification, or folding. However, the evolutionary mechanisms that drive cortical size and structure are unknown. Although genes that are essential for cortical developmental expansion have been identified from the genetics of human primary microcephaly (a disorder associated with reduced brain size and intellectual disability) 1 , studies of these genes in mice, which have a smooth cortex that is one thousand times smaller than the cortex of humans, have provided limited insight. Mutations in abnormal spindle-like microcephaly-associated (ASPM), the most common recessive microcephaly gene, reduce cortical volume by at least 50% in humans2-4, but have little effect on the brains of mice5-9; this probably reflects evolutionarily divergent functions of ASPM10,11. Here we used genome editing to create a germline knockout of Aspm in the ferret (Mustela putorius furo), a species with a larger, gyrified cortex and greater neural progenitor cell diversity12-14 than mice, and closer protein sequence homology to the human ASPM protein. Aspm knockout ferrets exhibit severe microcephaly (25-40% decreases in brain weight), reflecting reduced cortical surface area without significant change in cortical thickness, as has been found in human patients3,4, suggesting that loss of 'cortical units' has occurred. The cortex of fetal Aspm knockout ferrets displays a very large premature displacement of ventricular radial glial cells to the outer subventricular zone, where many resemble outer radial glia, a subtype of neural progenitor cells that are essentially absent in mice and have been implicated in cerebral cortical expansion in primates12-16. These data suggest an evolutionary mechanism by which ASPM regulates cortical expansion by controlling the affinity of ventricular radial glial cells for the ventricular surface, thus modulating the ratio of ventricular radial glial cells, the most undifferentiated cell type, to outer radial glia, a more differentiated progenitor.


Asunto(s)
Evolución Biológica , Corteza Cerebral/anatomía & histología , Corteza Cerebral/metabolismo , Hurones , Eliminación de Gen , Microcefalia/genética , Microcefalia/patología , Proteínas del Tejido Nervioso/deficiencia , Secuencia de Aminoácidos , Animales , Proteínas de Unión a Calmodulina/deficiencia , Proteínas de Unión a Calmodulina/metabolismo , Centrosoma/metabolismo , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Femenino , Hurones/anatomía & histología , Hurones/genética , Edición Génica , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Mutación de Línea Germinal , Humanos , Masculino , Ratones , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Tamaño de los Órganos , Transcripción Genética
8.
Neuron ; 92(4): 813-828, 2016 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-27974163

RESUMEN

Mutations in several genes encoding centrosomal proteins dramatically decrease the size of the human brain. We show that Aspm (abnormal spindle-like, microcephaly-associated) and Wdr62 (WD repeat-containing protein 62) interact genetically to control brain size, with mice lacking Wdr62, Aspm, or both showing gene dose-related centriole duplication defects that parallel the severity of the microcephaly and increased ectopic basal progenitors, suggesting premature delamination from the ventricular zone. Wdr62 and Aspm localize to the proximal end of the mother centriole and interact physically, with Wdr62 required for Aspm localization, and both proteins, as well as microcephaly protein Cep63, required to localize CENPJ/CPAP/Sas-4, a final common target. Unexpectedly, Aspm and Wdr62 are required for normal apical complex localization and apical epithelial structure, providing a plausible unifying mechanism for the premature delamination and precocious differentiation of progenitors. Together, our results reveal links among centrioles, apical proteins, and cell fate, and illuminate how alterations in these interactions can dynamically regulate brain size.


Asunto(s)
Encéfalo/embriología , Proteínas de Unión a Calmodulina/genética , Proteínas de Ciclo Celular/genética , Diferenciación Celular/genética , Centriolos/metabolismo , Microcefalia/genética , Proteínas Asociadas a Microtúbulos/genética , Proteínas del Tejido Nervioso/genética , Biogénesis de Organelos , Animales , Western Blotting , Encéfalo/metabolismo , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Inmunoprecipitación , Espectrometría de Masas , Ratones , Células Madre Embrionarias de Ratones , Mutación
9.
Cell ; 166(5): 1147-1162.e15, 2016 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-27565344

RESUMEN

Alternative splicing is prevalent in the mammalian brain. To interrogate the functional role of alternative splicing in neural development, we analyzed purified neural progenitor cells (NPCs) and neurons from developing cerebral cortices, revealing hundreds of differentially spliced exons that preferentially alter key protein domains-especially in cytoskeletal proteins-and can harbor disease-causing mutations. We show that Ptbp1 and Rbfox proteins antagonistically govern the NPC-to-neuron transition by regulating neuron-specific exons. Whereas Ptbp1 maintains apical progenitors partly through suppressing a poison exon of Flna in NPCs, Rbfox proteins promote neuronal differentiation by switching Ninein from a centrosomal splice form in NPCs to a non-centrosomal isoform in neurons. We further uncover an intronic human mutation within a PTBP1-binding site that disrupts normal skipping of the FLNA poison exon in NPCs and causes a brain-specific malformation. Our study indicates that dynamic control of alternative splicing governs cell fate in cerebral cortical development.


Asunto(s)
Empalme Alternativo , Corteza Cerebral/embriología , Células-Madre Neurales/citología , Neurogénesis/genética , Neuronas/citología , Animales , Centrosoma/metabolismo , Corteza Cerebral/anomalías , Corteza Cerebral/citología , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Exones , Ribonucleoproteínas Nucleares Heterogéneas/genética , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Humanos , Ratones , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína de Unión al Tracto de Polipirimidina/genética , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Dominios Proteicos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Factores de Empalme de ARN
10.
Elife ; 42015 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-26297806

RESUMEN

Primary microcephaly (MCPH) associated proteins CDK5RAP2, CEP152, WDR62 and CEP63 colocalize at the centrosome. We found that they interact to promote centriole duplication and form a hierarchy in which each is required to localize another to the centrosome, with CDK5RAP2 at the apex, and CEP152, WDR62 and CEP63 at sequentially lower positions. MCPH proteins interact with distinct centriolar satellite proteins; CDK5RAP2 interacts with SPAG5 and CEP72, CEP152 with CEP131, WDR62 with MOONRAKER, and CEP63 with CEP90 and CCDC14. These satellite proteins localize their cognate MCPH interactors to centrosomes and also promote centriole duplication. Consistent with a role for satellites in microcephaly, homozygous mutations in one satellite gene, CEP90, may cause MCPH. The satellite proteins, with the exception of CCDC14, and MCPH proteins promote centriole duplication by recruiting CDK2 to the centrosome. Thus, centriolar satellites build a MCPH complex critical for human neurodevelopment that promotes CDK2 centrosomal localization and centriole duplication.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Centriolos/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Línea Celular , Humanos , Multimerización de Proteína
11.
Neuron ; 84(6): 1240-57, 2014 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-25521379

RESUMEN

Katanin is a microtubule-severing complex whose catalytic activities are well characterized, but whose in vivo functions are incompletely understood. Human mutations in KATNB1, which encodes the noncatalytic regulatory p80 subunit of katanin, cause severe microlissencephaly. Loss of Katnb1 in mice confirms essential roles in neurogenesis and cell survival, while loss of zebrafish katnb1 reveals specific roles for katnin p80 in early and late developmental stages. Surprisingly, Katnb1 null mutant mouse embryos display hallmarks of aberrant Sonic hedgehog signaling, including holoprosencephaly. KATNB1-deficient human cells show defective proliferation and spindle structure, while Katnb1 null fibroblasts also demonstrate a remarkable excess of centrioles, with supernumerary cilia but deficient Hedgehog signaling. Our results reveal unexpected functions for KATNB1 in regulating overall centriole, mother centriole, and cilia number, and as an essential gene for normal Hedgehog signaling during neocortical development.


Asunto(s)
Adenosina Trifosfatasas/fisiología , Centriolos/fisiología , Corteza Cerebral/citología , Corteza Cerebral/embriología , Cilios/fisiología , Adenosina Trifosfatasas/genética , Animales , Estudios de Casos y Controles , Proliferación Celular/genética , Proliferación Celular/fisiología , Centriolos/genética , Corteza Cerebral/anomalías , Corteza Cerebral/metabolismo , Cilios/genética , Embrión de Mamíferos , Desarrollo Embrionario/genética , Fibroblastos/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Katanina , Ratones , Microcefalia/genética , Mutación , Linaje , Empalme del ARN/genética , Población Blanca/genética , Pez Cebra
12.
EMBO J ; 32(4): 597-607, 2013 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-23386061

RESUMEN

Formation of cilia, microtubule-based structures that function in propulsion and sensation, requires Kif3a, a subunit of Kinesin II essential for intraflagellar transport (IFT). We have found that, Kif3a is also required to organize centrioles. In the absence of Kif3a, the subdistal appendages of centrioles are disorganized and lack p150(Glued) and Ninein. Consequently, microtubule anchoring, centriole cohesion and basal foot formation are abrogated by loss of Kif3a. Kif3a localizes to the mother centriole and interacts with the Dynactin subunit p150(Glued). Depletion of p150(Glued) phenocopies the effects of loss of Kif3a, indicating that Kif3a recruitment of p150(Glued) is critical for subdistal appendage formation. The transport functions of Kif3a are dispensable for subdistal appendage organization as mutant forms of Kif3a lacking motor activity or the motor domain can restore p150(Glued) localization. Comparison to cells lacking Ift88 reveals that the centriolar functions of Kif3a are independent of IFT. Thus, in addition to its ciliogenic roles, Kif3a recruits p150(Glued) to the subdistal appendages of mother centrioles, critical for centrosomes to function as microtubule-organizing centres.


Asunto(s)
Centriolos/metabolismo , Cinesinas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Animales , Centriolos/genética , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Complejo Dinactina , Células HeLa , Humanos , Cinesinas/genética , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
13.
Mol Biol Cell ; 21(19): 3376-85, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20719959

RESUMEN

The centrosome contains proteins that control the organization of the microtubule cytoskeleton in interphase and mitosis. Its protein composition is tightly regulated through selective and cell cycle-dependent recruitment, retention, and removal of components. However, the mechanisms underlying protein delivery to the centrosome are not completely understood. We describe a novel function for the polarity protein Par6α in protein transport to the centrosome. We detected Par6α at the centrosome and centriolar satellites where it interacted with the centriolar satellite protein PCM-1 and the dynactin subunit p150(Glued). Depletion of Par6α caused the mislocalization of p150(Glued) and centrosomal components that are critical for microtubule anchoring at the centrosome. As a consequence, there were severe alterations in the organization of the microtubule cytoskeleton in the absence of Par6α and cell division was blocked. We propose a model in which Par6α controls centrosome organization through its association with the dynactin subunit p150(Glued).


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Autoantígenos/metabolismo , Proteínas de Ciclo Celular/metabolismo , Centrosoma/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Subunidades de Proteína/metabolismo , Línea Celular , Centriolos/metabolismo , Complejo Dinactina , Dineínas/metabolismo , Humanos , Interfase , Microtúbulos/metabolismo , Modelos Biológicos , Unión Proteica , Transporte de Proteínas
15.
Mol Biol Cell ; 20(4): 1192-200, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19109421

RESUMEN

The physical proximity of the Golgi apparatus and the centrosome is a unique feature of mammalian cells whose functional significance is only poorly understood. Here, we demonstrate that the previously described regulation of centrosome organization and function by the Golgi protein, GM130, involves a Golgi-associated complex consisting of GM130, the Rho GTPase, Cdc42, and its guanine nucleotide exchange factor, Tuba. We identified Tuba as a novel GM130-interacting protein and showed that this association controls Tuba-mediated activation of Cdc42 at the Golgi apparatus. Blocking either Tuba or Cdc42 activity reproduced the GM130 depletion phenotype of aberrant, nonfunctional centrosomes. Expression of constitutively active Cdc42 bypassed the requirement for GM130 in centrosome regulation, indicating that Cdc42 functions downstream of GM130. Our studies demonstrate that Cdc42 has a novel role in controlling centrosome organization in unstimulated cells in addition to its known function as a regulator of centrosome reorientation in stimulated cells. This first description of a regulatory pathway between the Golgi apparatus and the interphase centrosome that complements the known role of Golgi proteins in controlling spindle formation during mitosis and may provide an explanation for the pericentriolar position of the mammalian Golgi apparatus during interphase.


Asunto(s)
Autoantígenos/metabolismo , Centrosoma/enzimología , Aparato de Golgi/enzimología , Proteínas de la Membrana/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Proteínas del Citoesqueleto/metabolismo , Activación Enzimática , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células HeLa , Humanos , Interfase , Microtúbulos/enzimología , Mitosis , Unión Proteica
16.
Mol Biol Cell ; 19(2): 745-53, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18045989

RESUMEN

The Golgi apparatus (GA) of mammalian cells is positioned in the vicinity of the centrosome, the major microtubule organizing center of the cell. The significance of this physical proximity for organelle function and cell cycle progression is only beginning to being understood. We have identified a novel function for the GA protein, GM130, in the regulation of centrosome morphology, position and function during interphase. RNA interference-mediated depletion of GM130 from five human cell lines revealed abnormal interphase centrosomes that were mispositioned and defective with respect to microtubule organization and cell migration. When GM130-depleted cells entered mitosis, they formed multipolar spindles, arrested in metaphase, and died. We also detected aberrant centrosomes during interphase and multipolar spindles during mitosis in ldlG cells, which do not contain detectable GM130. Although GA proteins have been described to regulate mitotic centrosomes and spindle formation, this is the first report of a role for a GA protein in the regulation of centrosomes during interphase.


Asunto(s)
Autoantígenos/metabolismo , Centrosoma/metabolismo , Aparato de Golgi/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Autoantígenos/genética , Secuencia de Bases , Células CHO , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Centrosoma/patología , Cricetinae , Cricetulus , Fase G2 , Proteínas de la Matriz de Golgi , Humanos , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Microtúbulos/metabolismo , Mitosis , Datos de Secuencia Molecular , Huso Acromático/metabolismo , Huso Acromático/patología
17.
J Cell Sci ; 118(Pt 19): 4381-91, 2005 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16144865

RESUMEN

Phosphorylation of the membrane-cytoskeleton linker protein ezrin has been functionally linked to acid secretion and vesicle recruitment to the apical secretory membrane in gastric parietal cells. Phosphorylation of the conserved T567 residue of ezrin has been shown to alter the N/C oligomerization of ezrin and promote the formation of actin-rich surface projections in other cells. To test the importance of T567 as a regulatory site for ezrin in parietal cell activation, we incorporated wild-type (WT) and mutant forms of ezrin, including the nonphosphorylatable T567A mutation and a mutant mimicking permanent phosphorylation, T567D. All ezrin constructs included C-terminal cyan-fluorescent protein (CFP) and were incorporated into adenoviral constructs for efficient introduction into cultured parietal cells from rabbit stomach. Fluorescence microscopy was used to localize CFP-ezrin and monitor morphological responses. Accumulation of a weak base (aminopyrine) was used to monitor receptor-mediated acid secretory response of the cultured cells. Similar to endogenous ezrin, WT and T567A CFP-ezrin localized heavily to apical membrane vacuoles with considerably lower levels associated with the surrounding basolateral membrane. Interestingly, H,K-ATPase within cytoplasmic tubulovesicles was incorporated into the apical vacuoles along with WT and T567A mutant ezrin. In these parietal cells secretagogue stimulation produced a striking vacuolar expansion associated with HCl secretion and the secretory phenotype. Expression of T567D CFP-ezrin was quite different, being rarely associated with apical vacuoles. T567D was more typically localized to the basolateral membrane, often associated with long spikes and fingerlike projections. Moreover, the cells did not display secretagogue-dependent morphological changes and, to our surprise, H,K-ATPase was recruited to the T567D CFP-ezrin-enriched basolateral projections. We conclude that T567 phosphorylation, which is probably regulated through Rho signaling pathway, may direct ezrin to membrane-cytoskeletal activity at the basolateral membrane and away from apical secretory activity. The large basolateral expansion is predicted to recruit membranes from sources not normally targeted to that surface.


Asunto(s)
Polaridad Celular , Células Parietales Gástricas/citología , Células Parietales Gástricas/metabolismo , Fosfoproteínas/metabolismo , Treonina/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Animales , Extensiones de la Superficie Celular/metabolismo , Células Cultivadas , Proteínas del Citoesqueleto , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Fenotipo , Fosfoproteínas/genética , Fosforilación , Conejos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Vacuolas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...