Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
2.
Methods Cell Biol ; 175: 235-249, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36967143

RESUMO

Joubert syndrome (JS) is an autosomal recessive ciliopathy that mainly affects the morphogenesis of the cerebellum and brain stem. To date, mutations in at least 39 genes have been identified in JS; all these gene-encoding proteins are involved in the biogenesis of the primary cilium and centrioles. Recent studies using the mouse model carrying deleted or mutated JS-related genes exhibited cerebellar hypoplasia with a reduction in neurogenesis; however, investigating specific neuronal behaviors during their development in vivo remains challenging. Here, we describe an in vivo cerebellar electroporation technique that can be used to deliver plasmids carrying GFP and/or shRNAs into the major cerebellar cell type, granule neurons, from their progenitor state to their maturation in a spatiotemporal-specific manner. By combining this method with cerebellar immunostaining and EdU incorporation, these approaches enable the investigation of the cell-autonomous effect of JS-related genes in granule neuron progenitors, including the pathogenesis of ectopic neurons and the defects in neuronal differentiation. This approach provides information toward understanding the multifaceted roles of JS-related genes during cerebellar development in vivo.


Assuntos
Anormalidades Múltiplas , Anormalidades do Olho , Doenças Renais Císticas , Camundongos , Animais , Cerebelo/metabolismo , Cerebelo/patologia , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/metabolismo , Anormalidades Múltiplas/patologia , Doenças Renais Císticas/genética , Doenças Renais Císticas/metabolismo , Doenças Renais Císticas/patologia , Anormalidades do Olho/genética , Anormalidades do Olho/metabolismo , Anormalidades do Olho/patologia , Retina , Neurônios/metabolismo , Diferenciação Celular/genética , Proteínas , Proliferação de Células/genética , Eletroporação
3.
J Biomed Sci ; 29(1): 24, 2022 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-35365182

RESUMO

BACKGROUND: Metastasis is the major cause of morbidity and mortality in cancer that involves in multiple steps including epithelial-mesenchymal transition (EMT) process. Centrosome is an organelle that functions as the major microtubule organizing center (MTOC), and centrosome abnormalities are commonly correlated with tumor aggressiveness. However, the conclusive mechanisms indicating specific centrosomal proteins participated in tumor progression and metastasis remain largely unknown. METHODS: The expression levels of centriolar/centrosomal genes in various types of cancers were first examined by in silico analysis of the data derived from The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), and European Bioinformatics Institute (EBI) datasets. The expression of STIL (SCL/TAL1-interrupting locus) protein in clinical specimens was further assessed by Immunohistochemistry (IHC) analysis and the oncogenic roles of STIL in tumorigenesis were analyzed using in vitro and in vivo assays, including cell migration, invasion, xenograft tumor formation, and metastasis assays. The transcriptome differences between low- and high-STIL expression cells were analyzed by RNA-seq to uncover candidate genes involved in oncogenic pathways. The quantitative polymerase chain reaction (qPCR) and reporter assays were performed to confirm the results. The chromatin immunoprecipitation (ChIP)-qPCR assay was applied to demonstrate the binding of transcriptional factors to the promoter. RESULTS: The expression of STIL shows the most significant increase in lung and various other types of cancers, and is highly associated with patients' survival rate. Depletion of STIL inhibits tumor growth and metastasis. Interestingly, excess STIL activates the EMT pathway, and subsequently enhances cancer cell migration and invasion. Importantly, we reveal an unexpected role of STIL in tumor metastasis. A subset of STIL translocate into nucleus and associate with FOXM1 (Forkhead box protein M1) to promote tumor metastasis and stemness via FOXM1-mediated downstream target genes. Furthermore, we demonstrate that hypoxia-inducible factor 1α (HIF1α) directly binds to the STIL promoter and upregulates STIL expression under hypoxic condition. CONCLUSIONS: Our findings indicate that STIL promotes tumor metastasis through the HIF1α-STIL-FOXM1 axis, and highlight the importance of STIL as a promising therapeutic target for lung cancer treatment.


Assuntos
Transição Epitelial-Mesenquimal , Oncogenes , Movimento Celular/genética , Transição Epitelial-Mesenquimal/genética , Proteína Forkhead Box M1/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética
4.
Genes Dev ; 35(21-22): 1445-1460, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34711653

RESUMO

Joubert syndrome (JS) is a recessive ciliopathy in which all affected individuals have congenital cerebellar vermis hypoplasia. Here, we report that CEP120, a JS-associated protein involved in centriole biogenesis and cilia assembly, regulates timely neuronal differentiation and the departure of granule neuron progenitors (GNPs) from their germinal zone during cerebellar development. Our results show that depletion of Cep120 perturbs GNP cell cycle progression, resulting in a delay of cell cycle exit in vivo. To dissect the potential mechanism, we investigated the association between CEP120 interactome and the JS database and identified KIAA0753 (a JS-associated protein) as a CEP120-interacting protein. Surprisingly, we found that CEP120 recruits KIAA0753 to centrioles, and that loss of this interaction induces accumulation of GNPs in the germinal zone and impairs neuronal differentiation. Importantly, the replenishment of wild-type CEP120 rescues the above defects, whereas expression of JS-associated CEP120 mutants, which hinder KIAA0753 recruitment, does not. Together, our data reveal a close interplay between CEP120 and KIAA0753 for the germinal zone exit and timely neuronal differentiation of GNPs during cerebellar development, and mutations in CEP120 and KIAA0753 may participate in the heterotopia and cerebellar hypoplasia observed in JS patients.


Assuntos
Centríolos , Doenças Renais Císticas , Anormalidades Múltiplas , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Centríolos/genética , Centríolos/metabolismo , Cerebelo/anormalidades , Cerebelo/metabolismo , Anormalidades do Olho , Humanos , Doenças Renais Císticas/genética , Doenças Renais Císticas/metabolismo , Proteínas Associadas aos Microtúbulos , Retina/anormalidades
5.
Cells ; 10(6)2021 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-34207628

RESUMO

Autosomal recessive primary microcephaly (MCPH) is a complex neurodevelopmental disorder characterized by a small brain size with mild to moderate intellectual disability. We previously demonstrated that human microcephaly RTTN played an important role in regulating centriole duplication during interphase, but the role of RTTN in mitosis is not fully understood. Here, we show that RTTN is required for normal mitotic progression and correct spindle position. The depletion of RTTN induces the dispersion of the pericentriolar protein γ-tubulin and multiple mitotic abnormalities, including monopolar, abnormal bipolar, and multipolar spindles. Importantly, the loss of RTTN altered NuMA/p150Glued congression to the spindle poles, perturbed NuMA cortical localization, and reduced the number and the length of astral microtubules. Together, our results provide a new insight into how RTTN functions in mitosis.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Células Epiteliais , Microcefalia , Retina , Linhagem Celular , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Humanos , Microcefalia/metabolismo , Microcefalia/patologia , Mitose , Retina/citologia , Retina/metabolismo , Retina/patologia , Fuso Acromático/metabolismo
6.
Cell Death Differ ; 28(6): 1865-1879, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33462409

RESUMO

The DNA-PK maintains cell survival when DNA damage occurs. In addition, aberrant activation of the DNA-PK induces centrosome amplification, suggesting additional roles for this kinase. Here, we showed that the DNA-PK-p53 cascade induced primary cilia formation (ciliogenesis), thus maintaining the DNA damage response under genotoxic stress. Treatment with genotoxic drugs (etoposide, neocarzinostatin, hydroxyurea, or cisplatin) led to ciliogenesis in human retina (RPE1), trophoblast (HTR8), lung (A459), and mouse Leydig progenitor (TM3) cell lines. Upon genotoxic stress, several DNA damage signaling were activated, but only the DNA-PK-p53 cascade contributed to ciliogenesis, as pharmacological inhibition or genetic depletion of this pathway decreased genotoxic stress-induced ciliogenesis. Interestingly, in addition to localizing to the nucleus, activated DNA-PK localized to the base of the primary cilium (mother centriole) and daughter centriole. Genotoxic stress also induced autophagy. Inhibition of autophagy initiation or lysosomal degradation or depletion of ATG7 decreased genotoxic stress-induced ciliogenesis. Besides, inhibition of ciliogenesis by depletion of IFT88 or CEP164 attenuated the genotoxic stress-induced DNA damage response. Thus, our study uncovered the interplay among genotoxic stress, the primary cilium, and the DNA damage response.


Assuntos
Cílios/metabolismo , Dano ao DNA/genética , Proteína Quinase Ativada por DNA/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Autofagia , Humanos , Camundongos
7.
Sci Rep ; 9(1): 6037, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30988386

RESUMO

Centrosomal protein 120 (CEP120) was originally identified as a daughter centriole-enriched protein that participates in centriole elongation. Recent studies showed that CEP120 gene mutations cause complex ciliopathy phenotypes in humans, including Joubert syndrome and Jeune asphyxiating thoracic dystrophy, suggesting that CEP120 plays an additional role in ciliogenesis. To investigate the potential roles of CEP120 in centriole elongation and cilia formation, we knocked out the CEP120 gene in p53-deficient RPE1 cells using the CRISPR/Cas9 editing system, and performed various analyses. We herein report that loss of CEP120 produces short centrioles with no apparent distal and subdistal appendages. CEP120 knockout was also associated with defective centriole elongation, impaired recruitment of C2CD3 and Talpid3 to the distal ends of centrioles, and consequent defects in centriole appendage assembly and cilia formation. Interestingly, wild-type CEP120 interacts with C2CD3 and Talpid3, whereas a disease-associated CEP120 mutant (I975S) has a low affinity for C2CD3 binding and perturbs cilia assembly. Together, our findings reveal a novel role of CEP120 in ciliogenesis by showing that it interacts with C2CD3 and Talpid3 to assemble centriole appendages and by illuminating the molecular mechanism through which the CEP120 (I975S) mutation causes complex ciliopathies.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Centríolos/metabolismo , Cílios/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Sistemas CRISPR-Cas , Proteínas de Ciclo Celular/genética , Linhagem Celular , Centríolos/genética , Centríolos/ultraestrutura , Cílios/genética , Cílios/ultraestrutura , Ciliopatias/genética , Ciliopatias/metabolismo , Síndrome de Ellis-Van Creveld/genética , Síndrome de Ellis-Van Creveld/metabolismo , Deleção de Genes , Células HEK293 , Humanos , Mutação de Sentido Incorreto , Mapas de Interação de Proteínas
8.
Macromol Biosci ; 18(3)2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29231281

RESUMO

Neural tissue engineering has become a potential technology to restore the functionality of damaged neural tissue with the hope to cure the patients with neural disorder and to improve their quality of life. This paper reports the design and synthesis of polypeptides containing neuron stimulate, glutamic acid, for the fabrication of biomimetic 3D scaffold in neural tissue engineering application. The polypeptides are synthesized by efficient chemical reactions. Monomer γ-benzyl glutamate-N-carboxyanhydride undergoes ring-opening polymerization to form poly(γ-benzyl-l-glutamate), then hydrolyzes into poly(γ-benzyl-l-glutamate)-r-poly(glutamic acid) random copolymer. The glutamic acid amount is controlled by hydrolysis time. The obtained polymer molecular weight is in the range of 200 kDa for good quality of fibers. The fibrous 3D scaffolds of polypeptides are fabricated using electrospinning techniques. The scaffolds are biodegradable and biocompatible. The biocompatibility and length of neurite growth are improved with increasing amount of glutamic acid in scaffold. The 3D scaffold fabricated from aligned fibers can guide anisotropic growth of neurite along the fiber and into 3D domain. Furthermore, the length of neurite outgrowth is longer for scaffold made from aligned fibers as compared with that of isotropic fibers. This new polypeptide has potential for the application in the tissue engineering for neural regeneration.


Assuntos
Regeneração Nervosa , Ácido Poliglutâmico , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Materiais Biocompatíveis/química , Neurônios , Células PC12 , Ratos
9.
J Cell Sci ; 129(13): 2501-13, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27185865

RESUMO

Centriole duplication is a tightly ordered process during which procentrioles are assembled in G1-S and elongate during S and G2. Here, we show that human CEP295 (Drosophila Ana1) is not essential for initial cartwheel assembly, but is required to build distal half centrioles during S and G2. Using super-resolution and immunogold electron microscopy, we demonstrate that CEP295 is recruited to the proximal end of procentrioles in early S phase, when it is also localized at the centriolar microtubule wall that surrounds the human SAS6 cartwheel hub. Interestingly, depletion of CEP295 not only inhibits the recruitments of POC5 and POC1B to the distal half centrioles in G2, resulting in shorter centrioles, it also blocks the post-translational modification of centriolar microtubules (e.g. acetylation and glutamylation). Importantly, our results indicate that CEP295 directly interacts with microtubules, and that excess CEP295 could induce the assembly of overly long centrioles. Furthermore, exogenous expression of the N-terminal domain of CEP295 exerts a dominant-negative effect on centriole elongation. Collectively, these findings suggest that CEP295 is essential for building the distal half centrioles and for post-translational modification of centriolar microtubules.


Assuntos
Proteínas de Transporte/genética , Proteínas de Ciclo Celular/genética , Centríolos/genética , Microtúbulos/genética , Animais , Proteínas de Transporte/metabolismo , Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Centríolos/ultraestrutura , Centrossomo/metabolismo , Centrossomo/ultraestrutura , Regulação Neoplásica da Expressão Gênica , Células HeLa , Humanos , Microscopia Eletrônica , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Ligação Proteica/genética , Processamento de Proteína Pós-Traducional/genética
10.
Cell Rep ; 14(12): 2975-87, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-26997271

RESUMO

CPAP is required for centriole elongation during S/G2 phase, but the role of CPAP in mitosis is incompletely understood. Here, we show that CPAP maintains spindle pole integrity through its phosphorylation by Aurora-A during mitosis. Depletion of CPAP induced a prolonged delay in mitosis, pericentriolar material (PCM) dispersion, and multiple mitotic abnormalities. Further studies demonstrated that CPAP directly interacts with and is phosphorylated by Aurora-A at serine 467 during mitosis. Interestingly, the dispersal of the PCM was effectively rescued by ectopic expression of wild-type CPAP or a phospho-mimic CPAP-S467D mutant, but not a non-phosphorylated CPAP-S467A mutant. Finally, we found that CPAP-S467D has a low affinity for microtubule binding but a high affinity for PCM proteins. Together, our results support a model wherein CPAP is required for proper mitotic progression, and phosphorylation of CPAP by Aurora-A is essential for maintaining spindle pole integrity.


Assuntos
Aurora Quinase A/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Mitose , Polos do Fuso/fisiologia , Antígenos/genética , Antígenos/metabolismo , Aurora Quinase A/genética , Proteínas de Ciclo Celular , Centrossomo/fisiologia , Células HeLa , Humanos , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Microscopia Confocal , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Fosforilação , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Imagem com Lapso de Tempo , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
11.
Front Oncol ; 5: 178, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26322271

RESUMO

The meiotic generation of haploid gametes with equal contents of genetic material is important for sexual reproduction in mammals. Errors in the transmission of chromosomes during meiosis may lead to aneuploidy, which is the leading cause of miscarriage and congenital birth defects in humans. The Aurora kinases, which include Aurora-A, Aurora-B, and Aurora-C, are highly conserved serine-threonine kinases that play essential roles in centrosome function, chromosome segregation, and cytokinesis during mitosis and meiosis. While Aurora-A and Aurora-B have been extensively studied in mitosis, the role of Aurora-C in meiosis is only now starting to be revealed. For example, the perturbation of Aurora-C kinase activity by microinjection of Aurora-C-kinase-dead mutant mRNAs into mouse oocytes induced multiple defects, including chromosome misalignment, abnormal kinetochore-microtubule attachment, premature chromosome segregation, and failure of cytokinesis during meiotic division. However, the analysis of such defects is complicated by the possibility that Aurora-B may be present in mammalian germ cells. Interestingly, a homozygous mutation of Aurora-C in humans leads to the production of large-headed polyploid spermatozoa and causes male infertility, but homozygous females are fertile. Mouse studies regarding the roles of Aurora-B and Aurora-C in female meiotic divisions have yielded inconsistent results, and it has proven difficult to explain why homozygous human females have no significant clinical phenotype. In this review, we will discuss the controversial status of Aurora-B in oocytes and the possible role of Aurora-C during meiotic division.

12.
J Hepatol ; 58(6): 1157-64, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23369793

RESUMO

BACKGROUND & AIMS: Constitutive activation of NF-κB is an important event involved in chronic inflammation in hepatocellular carcinoma (HCC). CPAP, which plays important roles in centrosomal functions, was previously identified as the transcriptional co-activator of NF-κB. However, the molecular mechanism is unclear. The goal of this study was to investigate the role of CPAP in activating the NF-κB pathway in HCC. METHODS: SK-Hep1, HuH7, HepG2, HepG2X, Hep3B, and Hep3BX cells with CPAP overexpression or CPAP siRNA were used to evaluate activation of NF-κB under TNF-α stimulation by reporter assay, RT-PCR, Q-PCR, and Western blot analysis. In vivo SUMO modification of CPAP was demonstrated by an in situ PLA assay. Human HCC tissues were used to perform Q-PCR, Western blot, and IHC. RESULTS: CPAP siRNA abolished the interaction between IKKß and NF-κB, whereas overexpression of CPAP enhanced this interaction and finally led to augmented NF-κB activation by increasing the phosphorylation of NF-κB. CPAP could enter nuclei by associating with NF-κB. Furthermore, CPAP was SUMO-1 modified upon TNF-α stimulus, and this is essential for its NF-κB co-activator activity. SUMO-1-deficient CPAP mutant lost its NF-κB co-activator activity and failed to enter nuclei. Importantly, SUMOylated CPAP could synergistically increase the HBx-induced NF-κB activity. CONCLUSIONS: CPAP is essential for the recruitment of the IKK complex to inactivated NF-κB upon TNF-α treatment. Expression of CPAP was positively correlated with a poor prognosis in HBV-HCC. CPAP has the potential to serve as a therapeutic target for inflammation and inflammation-related diseases.


Assuntos
Carcinoma Hepatocelular/etiologia , Quinase I-kappa B/fisiologia , Neoplasias Hepáticas/etiologia , Proteínas Associadas aos Microtúbulos/fisiologia , NF-kappa B/fisiologia , Transdução de Sinais/fisiologia , Sumoilação , Transativadores/fisiologia , Carcinoma Hepatocelular/metabolismo , Humanos , Proteínas I-kappa B/metabolismo , Neoplasias Hepáticas/metabolismo , Inibidor de NF-kappaB alfa , Fosforilação , Proteína SUMO-1/fisiologia , Fator de Necrose Tumoral alfa/farmacologia , Proteínas Virais Reguladoras e Acessórias
13.
Mol Biol Cell ; 21(14): 2371-83, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20484572

RESUMO

We previously isolated Aurora-C/Aie1 in a screen for kinases expressed in mouse sperm and eggs. Here, we show the localization of endogenous Aurora-C and examine its roles during female mouse meiosis. Aurora-C was detected at the centromeres and along the chromosome arms in prometaphase I-metaphase I and was concentrated at centromeres at metaphase II, in which Aurora-C also was phosphorylated at Thr171. During the anaphase I-telophase I transition, Aurora-C was dephosphorylated and relocalized to the midzone and midbody. Microinjection of the kinase-deficient Aurora-C (AurC-KD) mRNA into mouse oocytes significantly inhibited Aurora-C activity and caused multiple defects, including chromosome misalignment, abnormal kinetochore-microtubule attachment, premature chromosome segregation, and cytokinesis failure in meiosis I. Furthermore, AurC-KD reduced Aurora-C and histone H3 phosphorylation and inhibited kinetochore localization of Bub1 and BubR1. Similar effects also were observed in the oocytes injected with INCNEP-delIN mRNAs, in which the Aurora-C binding motif was removed. The most dramatic effect observed in AurC-KD-injected oocytes is cytokinesis failure in meiosis I, resulting in producing large polyploid oocytes, a pattern similar to Aurora-C deficiency human spermatozoa. Surprisingly, we detected no Aurora-B protein in mouse oocytes. We propose that Aurora-C, but not Aurora-B, plays essential roles in female mouse meiosis.


Assuntos
Citocinese , Meiose , Oócitos/citologia , Oócitos/enzimologia , Poliploidia , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Aurora Quinase B , Aurora Quinase C , Aurora Quinases , Proteínas de Ciclo Celular , Proteínas Cromossômicas não Histona/metabolismo , Segregação de Cromossomos , Cromossomos de Mamíferos/metabolismo , Feminino , Proteínas de Fluorescência Verde/metabolismo , Histonas/metabolismo , Cinetocoros/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microtúbulos/metabolismo , Modelos Biológicos , Mutação/genética , Fosforilação , Fosfosserina/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Transporte Proteico
14.
Blood ; 114(19): 4233-42, 2009 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-19729518

RESUMO

The tightly regulated production of distinct erythrocyte protein 4.1R isoforms involves differential splicing of 3 mutually exclusive first exons (1A, 1B, 1C) to the alternative 3' splice sites (ss) of exon 2'/2. Here, we demonstrate that exon 1 and 2'/2 splicing diversity is regulated by a transcription-coupled splicing mechanism. We also implicate distinctive regulatory elements that promote the splicing of exon 1A to the distal 3' ss and exon 1B to the proximal 3' ss in murine erythroleukemia cells. A hybrid minigene driven by cytomegalovirus promoter mimicked 1B-promoter-driven splicing patterns but differed from 1A-promoter-driven splicing patterns, suggesting that promoter identity affects exon 2'/2 splicing. Furthermore, splicing factor SF2/ASF ultraviolet (UV) cross-linked to the exon 2'/2 junction CAGAGAA, a sequence that overlaps the distal U2AF(35)-binding 3' ss. Consequently, depletion of SF2/ASF allowed exon 1B to splice to the distal 3' ss but had no effect on exon 1A splicing. These findings identify for the first time that an SF2/ASF binding site also can serve as a 3' ss in a transcript-dependent manner. Taken together, our results suggest that 4.1R gene expression involves transcriptional regulation coupled with a complex splicing regulatory network.


Assuntos
Processamento Alternativo , Proteínas Sanguíneas/genética , Proteínas do Citoesqueleto/genética , Proteínas de Membrana/genética , Regiões 5' não Traduzidas , Animais , Sequência de Bases , Sítios de Ligação/genética , Proteínas Sanguíneas/biossíntese , Linhagem Celular , Proteínas do Citoesqueleto/biossíntese , DNA Polimerase II/metabolismo , Primers do DNA/genética , Éxons , Humanos , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/metabolismo , Proteínas de Membrana/biossíntese , Camundongos , Proteínas dos Microfilamentos , Modelos Genéticos , Dados de Sequência Molecular , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteínas/metabolismo , Fatores de Processamento de Serina-Arginina , Fator de Processamento U2AF , Distribuição Tecidual , Transcrição Gênica , Ativação Transcricional , Células Tumorais Cultivadas
15.
Nat Cell Biol ; 11(7): 825-31, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19503075

RESUMO

Centriole duplication involves the growing of a procentriole (progeny centriole) next to the proximal end of each pre-existing centriole (parental centriole). The molecular mechanisms that regulate procentriole elongation remain obscure. We show here that expression of the centriolar protein CPAP (centrosomal P4.1-associated protein) is carefully regulated during the cell cycle, with the protein being degraded in late mitosis. Depletion of CPAP inhibited centrosome duplication, whereas excess CPAP induced the formation of elongated procentriole-like structures (PLSs), which contain stable microtubules and several centriolar proteins. Ultrastructural analysis revealed that these structures are similar to procentrioles with elongated microtubules. Overexpression of a CPAP mutant (CPAP-377EE) that does not bind to tubulin dimers significantly inhibited the formation of CPAP-induced PLSs. Together, these results suggest that CPAP is a new regulator of centriole length and its intrinsic tubulin-dimer binding activity is required for procentriole elongation.


Assuntos
Ciclo Celular/fisiologia , Centríolos/metabolismo , Proteínas Associadas aos Microtúbulos/fisiologia , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Citometria de Fluxo , Células HeLa , Humanos , Immunoblotting , Imunoprecipitação , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Mitose/genética , Mitose/fisiologia , Modelos Biológicos , RNA Interferente Pequeno , Tubulina (Proteína)/metabolismo , Ubiquitinação
16.
Exp Cell Res ; 314(14): 2591-602, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18586240

RESUMO

We previously identified a novel centrosomal protein CPAP, which carries a 112-residue motif that is essential for microtubule destabilization. In this report, we define both the microtubule (MT) binding and destabilizing domains in human CPAP and analyze the mutations that affect its MT-destabilizing activity. Analysis of a series of CPAP truncated proteins showed that the MT-binding domain (MBD; residues 423-607) of CPAP is located next to its MT-destabilizing domain (MDD; residues 311-422). Site-specific mutagenesis revealed that the mutations that either disrupt the alpha-helical structure (Y341P, I346P, L348P, and triple-P) or alter the charge property (KR377EE) of the MDD significantly affect its MT-destabilizing ability. The activity for binding to a tubulin heterodimer was also significantly reduced in KR377EE mutant. Furthermore, we have analyzed the putative function of Drosophila d-SAS-4, a distant relative of human CPAP, which shares a conserved approximately 20-aa sequence with the MDD of CPAP. Our results show that mutations in this conserved sequence also eliminate d-SAS-4's MT-destabilizing activity, suggesting that d-SAS-4 and CPAP may play similar roles within cells.


Assuntos
Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas Associadas aos Microtúbulos/química , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Sequência de Aminoácidos , Animais , Bovinos , Linhagem Celular Tumoral , Análise Mutacional de DNA , Dimerização , Proteínas de Fluorescência Verde/metabolismo , Humanos , Microtúbulos/efeitos dos fármacos , Dados de Sequência Molecular , Proteínas Mutantes/metabolismo , Paclitaxel/farmacologia , Peptídeos/metabolismo , Fenótipo , Ligação Proteica/efeitos dos fármacos , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos , Tubulina (Proteína)/metabolismo
17.
J Biomed Sci ; 12(2): 297-310, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15917996

RESUMO

Aurora kinases are emerging as key regulators of centrosome function, chromosome segregation and cytokinesis. We previously isolated Aurora-C (Aie1), a third type of Aurora kinase, in a screen for kinases expressed in mouse sperm and eggs. Currently, we know very little about the precise localization and function of Aurora-C. Immunofluorescence analysis of ectopically expressed GFP-Aurora-C has revealed that Aurora-C is a new member of the chromosomal passenger proteins localizing first to the centromeres and then to the central spindles during cytokinesis. In order to study the potential role of Aurora-C, we examined the effects of a kinase-deficient (KD) mutant (AurC-KD) in HeLa Tet-Off cells under tetracycline control. Our results showed that overexpression of AurC-KD causes defects in cell division and induces polyploidy and apoptosis. Interestingly, AurC-KD overexpression also inhibits centromere/kinetochore localization of Aurora-B, Bub1, and BubR1, reduces histone H3 phosphorylation, and disrupts the association of INCENP with Aurora-B. Together, our results showed that Aurora-C is a chromosomal passenger protein, which may serve as a key regulator in cell division.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Apoptose , Aurora Quinase B , Aurora Quinase C , Aurora Quinases , Proteínas de Ciclo Celular , Divisão Celular , Proliferação de Células , Separação Celular , Centrômero/metabolismo , Centrossomo/ultraestrutura , Citocinese , DNA Complementar/metabolismo , Citometria de Fluxo , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Histonas/metabolismo , Humanos , Immunoblotting , Marcação In Situ das Extremidades Cortadas , Substâncias Macromoleculares , Microscopia de Fluorescência , Mutagênese , Mutação , Fosforilação , Poliploidia , Ligação Proteica , Proteínas Quinases , Fuso Acromático , Tetraciclina/farmacologia , Fatores de Tempo , Transfecção
18.
Mol Endocrinol ; 16(9): 2019-33, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12198240

RESUMO

Stat5, a member of the signal transducer and activators of transcription (Stat) protein family, is a primary mediator of prolactin (PRL) signaling in the mammary gland. There are two distinct Stat5 genes, Stat5a and Stat5b. The Stat5a isoform has been demonstrated to have an essential role in mammary epithelial differentiation, whereas Stat5b is required for dimorphic sexual growth. To search for proteins that interact with the C terminus of Stat5a, a highly divergent region amongst Stat family members, we performed a yeast two-hybrid screen of HBL100 and primary breast adenocarcinoma libraries. This led to the identification of a protein that had previously been isolated as a centrosomal P4.1-associated protein (CPAP). CPAP was shown to specifically interact with Stat5a and Stat5b but not with Stat1 or Stat3. Both the tyrosine phosphorylated and unphosphorylated forms of Stat5, as well as Stat5a/Stat5b heterodimers, could associate with CPAP. CPAP was expressed in human breast cancer cell lines and the developing mammary gland as well as in other tissues. Indirect immunofluorescence and cellular fractionation studies revealed that CPAP was predominantly cytoplasmic, with low levels in the nucleus. Nuclear levels of CPAP increased substantially upon activation of the PRL pathway, most likely reflecting cotranslocation of this protein with activated Stat5. Furthermore, CPAP was found to augment Stat5-mediated transcription. Thus, we have identified CPAP as a novel coactivator of Stat5 proteins in the PRL (and probably other) pathways.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Leite , Transativadores/metabolismo , Transcrição Gênica , Transporte Ativo do Núcleo Celular , Animais , Sítios de Ligação , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/química , Perfilação da Expressão Gênica , Humanos , Substâncias Macromoleculares , Camundongos , Especificidade de Órgãos , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT5 , Transativadores/química , Ativação Transcricional , Proteínas Supressoras de Tumor , Técnicas do Sistema de Duplo-Híbrido
19.
DNA Cell Biol ; 21(1): 41-6, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11879579

RESUMO

We previously reported two novel serine/threonine kinases, Aie1 (mouse) and AIE2 (human), both later referred to as aurora-C, a newly recognized member of the mitotic aurora kinase family. In the present study, we analyzed the phosphorylation sites of mouse Aie1 by site-directed mutagenesis. Our results showed that protein kinase A (PKA) phosphorylates Aie1 at a threonine residue located at amino acid position 171. The T171A and T175A mutants, in which threonines located at residues 171 and 175 were replaced by alanines, revealed a significant increase in their kinase activities to phosphorylate ACS-1 (Aurora-C substrate 1). In contrast, the double mutant T171A-T175A showed impaired kinase activity. In addition, we had previously identified a PEST-like motif located at the N terminus of Aie1. Mutation analysis in the present study revealed that the quadruple mutant in which the PEST-like motif was mutated significantly abrogated Aie1 kinase activity. This is the first report of the analysis of potential phosphorylation sites of mouse aurora-C in vitro.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas Serina-Treonina Quinases/genética , Animais , Aurora Quinase C , Aurora Quinases , Sítios de Ligação , Análise Mutacional de DNA , Primers do DNA/química , Expressão Gênica , Técnicas In Vitro , Camundongos , Mutagênese Sítio-Dirigida , Fragmentos de Peptídeos , Fosforilação , Proteínas Serina-Treonina Quinases/isolamento & purificação , Proteínas Serina-Treonina Quinases/metabolismo , Treonina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA