Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 294(44): 16034-16048, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31506300

RESUMO

Sonic hedgehog (SHH) is important for organogenesis during development. Recent studies have indicated that SHH is also involved in the proliferation and transformation of astrocytes to the reactive phenotype. However, the mechanisms underlying these are unknown. Involvement of SHH signaling in calcium (Ca) signaling has not been extensively studied. Here, we report that SHH and Smoothened agonist (SAG), an activator of the signaling receptor Smoothened (SMO) in the SHH pathway, activate Ca oscillations in cultured murine hippocampal astrocytes. The response was rapid, on a minute time scale, indicating a noncanonical pathway activity. Pertussis toxin blocked the SAG effect, indicating an involvement of a Gi coupled to SMO. Depletion of extracellular ATP by apyrase, an ATP-degrading enzyme, inhibited the SAG-mediated activation of Ca oscillations. These results indicate that SAG increases extracellular ATP levels by activating ATP release from astrocytes, resulting in Ca oscillation activation. We hypothesize that SHH activates SMO-coupled Gi in astrocytes, causing ATP release and activation of Gq/11-coupled P2 receptors on the same cell or surrounding astrocytes. Transcription factor activities are often modulated by Ca patterns; therefore, SHH signaling may trigger changes in astrocytes by activating Ca oscillations. This enhancement of Ca oscillations by SHH signaling may occur in astrocytes in the brain in vivo because we also observed it in hippocampal brain slices. In summary, SHH and SAG enhance Ca oscillations in hippocampal astrocytes, Gi mediates SAG-induced Ca oscillations downstream of SMO, and ATP-permeable channels may promote the ATP release that activates Ca oscillations in astrocytes.


Assuntos
Astrócitos/metabolismo , Sinalização do Cálcio , Proteínas Hedgehog/metabolismo , Hipocampo/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Proteínas de Transporte/metabolismo , Células Cultivadas , Hipocampo/citologia , Camundongos , Camundongos Endogâmicos ICR , Receptor Smoothened/metabolismo
2.
Exp Cell Res ; 353(2): 79-87, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28284839

RESUMO

Chromosome instability, frequently found in cancer cells, is caused by a deficiency in cell division, including centrosomal amplification and cytokinesis failure, and can result in abnormal chromosome content or aneuploidy. The small GTPase pathways have been implicated as important processes in cell division. We found that knockdown of a tumor suppressor protein Kank1 increases the number of cells with a micronucleus or bi-/multi-nuclei, which was likely caused by centrosomal amplification. Kank1 interacts with Daam1, known to bind to and activate a small GTPase, RhoA, in actin assembly. Knockdown of Kank1 or overexpression of Daam1, respectively, hyperactivates RhoA, potentially leading to the modulation of the activity of Aurora-A, a key regulator of centrosomal functions, eventually resulting in centrosomal amplification. Kank1 is also associated with contractile ring formation in collaboration with RhoA, and its deficiency results in the interruption of normal daughter cell separation, generating multinucleate cells. Such abnormal segregation of chromosomes may cause further chromosomal instability and abnormal gene functions, leading to tumorigenesis. Thus, Kank1 plays a crucial role in regulating the activity of RhoA through retrieving excess Daam1 and balancing the activities of RhoA and its effectors.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Carcinogênese/genética , Neoplasias/genética , Proteínas Supressoras de Tumor/genética , Proteína rhoA de Ligação ao GTP/genética , Animais , Aurora Quinase A/genética , Divisão Celular/genética , Centrossomo/metabolismo , Instabilidade Cromossômica/genética , Segregação de Cromossomos/genética , Proteínas do Citoesqueleto , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Camundongos , Proteínas dos Microfilamentos , Células NIH 3T3 , Neoplasias/patologia , Proteínas rho de Ligação ao GTP
3.
Epilepsy Res ; 108(3): 420-32, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24480790

RESUMO

Mutations in GABRG2, which encodes the γ2 subunit of GABAA receptors, can cause both genetic epilepsy with febrile seizures plus (GEFS+) and Dravet syndrome. Most GABRG2 truncating mutations associated with Dravet syndrome result in premature termination codons (PTCs) and are stably translated into mutant proteins with potential dominant-negative effects. This study involved search for mutations in candidate genes for Dravet syndrome, namely SCN1A, 2A, 1B, 2B, GABRA1, B2, and G2. A heterozygous nonsense mutation (c.118C>T, p.Q40X) in GABRG2 was identified in dizygotic twin girls with Dravet syndrome and their apparently healthy father. Electrophysiological studies with the reconstituted GABAA receptors in HEK cells showed reduced GABA-induced currents when mutated γ2 DNA was cotransfected with wild-type α1 and ß2 subunits. In this case, immunohistochemistry using antibodies to the α1 and γ2 subunits of GABAA receptor showed granular staining in the soma. In addition, microinjection of mutated γ2 subunit cDNA into HEK cells severely inhibited intracellular trafficking of GABAA receptor subunits α1 and ß2, and retention of these proteins in the endoplasmic reticulum. The mutated γ2 subunit-expressing neurons also showed impaired axonal transport of the α1 and ß2 subunits. Our findings suggested that different phenotypes of epilepsy, e.g., GEFS+ and Dravet syndrome (which share similar abnormalities in causative genes) are likely due to impaired axonal transport associated with the dominant-negative effects of GABRG2.


Assuntos
Códon sem Sentido/genética , Epilepsia/genética , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Células Cultivadas , Pré-Escolar , Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/patologia , Feminino , Regulação da Expressão Gênica , Células HEK293 , Hipocampo/citologia , Humanos , Lactente , Japão , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Camundongos , Modelos Moleculares , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Subunidades Proteicas/genética , Transporte Proteico/genética , Frações Subcelulares/metabolismo , Frações Subcelulares/ultraestrutura , Gêmeos Dizigóticos
4.
Traffic ; 11(2): 287-301, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19958467

RESUMO

Functional defects in cilia are associated with various human diseases including congenital hydrocephalus. Previous studies suggested that defects in cilia not only disrupt the flow of cerebrospinal fluid (CSF) generated by motile cilia in ependyma lining the brain ventricles, but also cause increased CSF production at the choroid plexus. However, the molecular mechanisms of CSF overproduction by ciliary dysfunction remain elusive. To dissect the molecular mechanisms, choroid plexus epithelial cells (CPECs) were isolated from porcine brain. These cells expressed clusters of primary cilia on the apical surface. Deciliation of CPECs elevated the intracellular cyclic AMP (cAMP) levels and stimulated basolateral-to-apical fluid transcytosis, without detrimental effects on other morphological and physiological features. The primary cilia possessed neuropeptide FF (NPFF) receptor 2. In deciliated cells, the responsiveness to NPFF was reduced at nanomolar concentrations. Furthermore, CPECs expressed NPFF precursor along with NPFFR2. An NPFFR antagonist, BIBP3226, increased the fluid transcytosis, suggesting the presence of autocrine NPFF signaling in CPECs for a tonic inhibition of fluid transcytosis. These results suggest that the clusters of primary cilia in CPECs act as a sensitive chemosensor to regulate CSF production.


Assuntos
Cílios/metabolismo , Epitélio/metabolismo , Animais , Ansiolíticos/farmacologia , Arginina/análogos & derivados , Arginina/farmacologia , Comunicação Autócrina/efeitos dos fármacos , Sequência de Bases , Células CACO-2 , Bovinos , Linhagem Celular Tumoral , Líquido Cefalorraquidiano/metabolismo , Plexo Corióideo/citologia , Plexo Corióideo/metabolismo , Cílios/ultraestrutura , AMP Cíclico/metabolismo , Humanos , Camundongos , Modelos Biológicos , Dados de Sequência Molecular , Oligopeptídeos/metabolismo , Transporte Proteico/efeitos dos fármacos , Ratos , Receptores de Neuropeptídeos/metabolismo , Suínos
5.
Biochem Biophys Res Commun ; 386(4): 639-44, 2009 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-19559006

RESUMO

Congenital fibrosis of the extraocular muscles type 1 (CFEOM1) is associated with heterozygous mutations in the KIF21A gene, including a major (R954W) and a minor (M947T) mutation. Kank1, which regulates actin polymerization, cell migration and neurite outgrowth, interacted with the third and fourth coiled-coil domains of KIF21A protein at its ankyrin-repeat domain. While both KIF21A(R954W) and KIF21A(M947T) enhanced the formation of a heterodimer with the wild type, KIF21A(WT), these mutants also enhanced the interaction with Kank1. Knockdown of KIF21A resulted in Kank1 predominantly occurring in the cytosolic fraction, while KIF21A(WT) slightly enhanced the translocation of Kank1 to the membrane fraction. Moreover, KIF21A(R954W) significantly enhanced the translocation of Kank1 to the membrane fraction. These results suggest that KIF21A regulates the distribution of Kank1 and that KIF21A mutations associated with CFEOM1 enhanced the accumulation of Kank1 in the membrane fraction. This might cause an abrogation of neuronal development in cases of CFEOM1 through over-regulation of actin polymerization by Kank1.


Assuntos
Blefaroptose/congênito , Cinesinas/genética , Músculos Oculomotores/patologia , Oftalmoplegia/congênito , Proteínas Supressoras de Tumor/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Repetição de Anquirina/genética , Blefaroptose/genética , Blefaroptose/metabolismo , Membrana Celular/metabolismo , Proteínas do Citoesqueleto , Fibrose , Células HeLa , Humanos , Cinesinas/química , Cinesinas/metabolismo , Mutação , Oftalmoplegia/genética , Oftalmoplegia/metabolismo , Multimerização Proteica , Transporte Proteico , Proteínas Supressoras de Tumor/genética
6.
J Cell Biol ; 184(2): 253-67, 2009 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-19171758

RESUMO

In this study, insulin receptor substrate (IRS) p53 is identified as a binding partner for Kank, a kidney ankyrin repeat-containing protein that functions to suppress cell proliferation and regulate the actin cytoskeleton. Kank specifically inhibits the binding of IRSp53 with active Rac1 (Rac1(G12V)) but not Cdc42 (cdc42(G12V)) and thus inhibits the IRSp53-dependent development of lamellipodia without affecting the formation of filopodia. Knockdown (KD) of Kank by RNA interference results in increased lamellipodial development, whereas KD of both Kank and IRSp53 has little effect. Moreover, insulin-induced membrane ruffling is inhibited by overexpression of Kank. Kank also suppresses integrin-dependent cell spreading and IRSp53-induced neurite outgrowth. Our results demonstrate that Kank negatively regulates the formation of lamellipodia by inhibiting the interaction between Rac1 and IRSp53.


Assuntos
Actinas/metabolismo , Proteínas de Transporte/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte/genética , Células Cultivadas , Proteínas do Citoesqueleto , Células HeLa , Humanos , Camundongos , Pseudópodes/metabolismo , Interferência de RNA
7.
J Cell Biol ; 181(3): 537-49, 2008 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-18458160

RESUMO

Phosphoinositide-3 kinase (PI3K)/Akt signaling is activated by growth factors such as insulin and epidermal growth factor (EGF) and regulates several functions such as cell cycling, apoptosis, cell growth, and cell migration. Here, we find that Kank is an Akt substrate located downstream of PI3K and a 14-3-3-binding protein. The interaction between Kank and 14-3-3 is regulated by insulin and EGF and is mediated through phosphorylation of Kank by Akt. In NIH3T3 cells expressing Kank, the amount of actin stress fibers is reduced, and the coexpression of 14-3-3 disrupted this effect. Kank also inhibits insulin-induced cell migration via 14-3-3 binding. Furthermore, Kank inhibits insulin and active Akt-dependent activation of RhoA through binding to 14-3-3. Based on these findings, we hypothesize that Kank negatively regulates the formation of actin stress fibers and cell migration through the inhibition of RhoA activity, which is controlled by binding of Kank to 14-3-3 in PI3K-Akt signaling.


Assuntos
Proteínas 14-3-3/metabolismo , Movimento Celular/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia , Fibras de Estresse/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Proteínas 14-3-3/genética , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Aminoácidos , Animais , Repetição de Anquirina , Proteínas do Citoesqueleto , Ativação Enzimática , Células HeLa , Humanos , Insulina/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Dados de Sequência Molecular , Células NIH 3T3 , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Supressoras de Tumor/genética , Proteína rhoA de Ligação ao GTP/genética
8.
Biochim Biophys Acta ; 1780(2): 128-33, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17996375

RESUMO

The human Kank gene was found as a candidate tumor suppressor for renal cell carcinoma, and encodes an ankyrin-repeat domain-containing protein, Kank. Here, we report a new family of proteins consisting of three Kank (Kank1)-associated members, Kank2, Kank3 and Kank4, which were found by domain and phylogenetic analyses. Besides the conserved ankyrin-repeat and coiled-coil domains, there was a conserved motif at the N-terminal (KN motif) containing potential motifs for nuclear localization and export signals. Gene expression of these genes was examined by RT-PCR at the mRNA level and by Western blotting and immunostaining at the protein level. Kank family genes showed variations in the expression level among tissues and kidney cell lines. Furthermore, the results of overexpression of these genes in NIH3T3 cells suggest that all of these family proteins have an identical role in the formation of actin stress fibers.


Assuntos
Repetição de Anquirina , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/classificação , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Aminoácidos , Animais , Proteínas do Citoesqueleto , Expressão Gênica , Genoma , Humanos , Camundongos , Dados de Sequência Molecular , Células NIH 3T3 , Filogenia , Ratos , Proteínas Supressoras de Tumor/genética
9.
J Cell Sci ; 119(Pt 19): 4002-10, 2006 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16968744

RESUMO

The human Kank protein has a role in controlling the formation of the cytoskeleton by regulating actin polymerization. Besides the cytoplasmic localization as reported before, we observed the nuclear localization of Kank in OS-RC-2 cells. To uncover the mechanism behind this phenomenon, we focused on the nuclear localization signal (NLS) and the nuclear export signal (NES). We found one NLS (NLS1) and two NESs (NES1 and NES2) in the N-terminal region of Kank-L that were absent in Kank-S, and another NLS (NLS2) and NES (NES3) in the common region. These signals were active as mutations introduced into them abolished the nuclear import (for NLS1 and NLS2) or the nuclear export (for NES1 to NES3) of Kank. The localization of Kank in the cells before and after treatment with leptomycin B suggested that the transportation of Kank from the nucleus to the cytoplasm was mediated by a CRM1-dependent mechanism. TOPFLASH reporter assays revealed a positive relationship between the nuclear import of Kank and the activation of beta-catenin-dependent transcription. Kank can bind to beta-catenin and regulate the subcellular distribution of beta-catenin. Based on the findings shown here, we propose that Kank has multiple functions in the cells and plays different roles in the cytoplasm and the nucleus.


Assuntos
Transporte Ativo do Núcleo Celular/fisiologia , Transporte Proteico/fisiologia , Proteínas Supressoras de Tumor/metabolismo , beta Catenina/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal , Animais , Células Cultivadas , Proteínas do Citoesqueleto , Ácidos Graxos Insaturados/farmacologia , Humanos , Camundongos , Células NIH 3T3 , Sinais de Localização Nuclear/química , Sinais Direcionadores de Proteínas , Distribuição Tecidual/efeitos dos fármacos , Transcrição Gênica , Proteínas Supressoras de Tumor/química
10.
Biochem Biophys Res Commun ; 330(4): 1247-53, 2005 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-15823577

RESUMO

The human Kank gene encodes an ankyrin repeat domain-containing protein which regulates actin polymerization. There are at least two types of Kank protein depending on cell type, likely due to differences in transcription. Here, to examine the transcriptional initiation and genomic organization of the human Kank gene, we performed 5'-RACE (rapid amplification of cDNA ends) using total RNA from normal kidney and a human kidney cancer cell line, VMRC-RCW cells. The results suggest that the human Kank gene has several alternative first exons. While mRNA from VMRC-RCW cells encoded Kank protein (referred to as Kank-S) as reported previously, mRNA from the normal kidney tissue encoded a novel type of Kank protein (referred to as Kank-L), which contained an additional N-terminal sequence 158 amino acids long. Promoter activity and the expression of the Kank variants in normal and cancer tissues were examined.


Assuntos
Processamento Alternativo , Proteínas Supressoras de Tumor/genética , Proteínas Adaptadoras de Transdução de Sinal , Linhagem Celular , Linhagem Celular Tumoral , Biologia Computacional , Proteínas do Citoesqueleto , Éxons , Genes Reporter , Humanos , Rim/metabolismo , Neoplasias Renais/metabolismo , Luciferases/genética , Regiões Promotoras Genéticas , Proteínas Supressoras de Tumor/metabolismo
11.
Cancer Sci ; 96(3): 191-6, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15771623

RESUMO

The improved IGCR (In-Gel Competitive Reassociation) method was applied to the analysis of human gastric cancer genomic DNA to identify its alterations, and it appeared that the IGCR library contained a fragment of 3'-untranslated region (3' UTR) of G-protein coupled receptor 30 (GPR30) mRNA. When we searched genomic DNA pairs of gastric cancer patients with this IGCR clone, we found the deletion polymorphism with or without 2 bp (Cytosine and Thymine; CT). We confirmed the existence of a novel mRNA in GPR30 3'UTR by northern blotting, cloned this novel mRNA and named it Leucine Rich Protein in GPR30 3'UTR (LERGU). The EST database search gave one alternative splicing form in this 3' UTR, which was named as LERGU-1. A novel alternative splicing form of this mRNA was also identified from the stomach total RNA, which was named LERGU-2. The LERGU mRNA was also detected in eight gastric cancer cell lines, but GPR30 mRNA scarcely existed. Furthermore, we detected the 2 bp-deletion form in genomic DNAs and mRNAs derived from gastric cancers, but not in other type cancers. Since the 2 bp-deletion position on LERGU corresponds to its alternative splicing site, this deletion may produce a frame-shifted protein. Overall, our findings suggest that a mutation or disappearance of the normal LERGU protein may have a function in the development of gastric cancer.


Assuntos
DNA de Neoplasias/genética , Polimorfismo Genético , Receptores Acoplados a Proteínas G/genética , Neoplasias Gástricas/genética , Regiões 3' não Traduzidas , Sequência de Aminoácidos , Sequência de Bases , Clonagem Molecular , DNA de Neoplasias/análise , Mutação da Fase de Leitura , Humanos , Leucina , Dados de Sequência Molecular , RNA Mensageiro/análise , Receptores de Estrogênio , Neoplasias Gástricas/patologia
12.
Cancer Lett ; 213(1): 111-6, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15312690

RESUMO

Loss of heterozygosity (LOH) is an important event of tumorigenesis. In this paper, we report the comprehensive LOH analyses with microsatellite markers and their results at chromosome 8p11.1-q13.3 in gastric cancer. The microsatellite markers D8S2323 and D8S2330 exhibited high LOH frequencies, 54.2 and 57.1%, respectively. However, LOH at 8q showed no relationship to either histological types or stages of gastric cancer. Finally, we settled six candidate regions on 8q in gastric cancer where there was a high possibility of being the tumor suppressor gene(s), and concluded that the LOH of 8q occurred in the primary tumorigenesis of gastric cancer.


Assuntos
Cromossomos Humanos Par 8 , Genes Supressores de Tumor , Perda de Heterozigosidade , Neoplasias Gástricas/genética , Humanos , Repetições de Microssatélites , Reação em Cadeia da Polimerase , Neoplasias Gástricas/etiologia
13.
Int J Cancer ; 109(1): 71-5, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14735470

RESUMO

Loss of heterozygosity (LOH) is an important event of tumorigenesis. In gastric cancer, we found a novel region of LOH in chromosome 9q having about 800 kb deletions at 9q31.1. The microsatellite marker D9S938 in that region exhibiting the highest LOH frequency, 56.5%. In addition, the LOH at 9q31.1 did not show any relationship to either histologic types or stages of gastric cancers, and several genes were predicted in the remaining allele by in silico methods. These data suggest that the deletion at 9q31.1 would be common in both differentiated-type and undifferentiated-type gastric cancers. Furthermore, this deletion was found in the primary tumors of early-stage gastric cancer, indicating that loss of function of predicted genes appears to be associated with the tumorigenesis of gastric cancer.


Assuntos
Cromossomos Humanos Par 9/ultraestrutura , Genes Supressores de Tumor , Neoplasias Gástricas/genética , Diferenciação Celular , Mapeamento Cromossômico , Primers do DNA/farmacologia , Deleção de Genes , Genoma , Humanos , Perda de Heterozigosidade , Repetições de Microssatélites , Modelos Genéticos , RNA Mensageiro/metabolismo , Software
14.
Med Electron Microsc ; 32(4): 221-225, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11810449

RESUMO

It has been suggested that the LGN protein is associated with Galphai2 by the yeast two-hybrid system and in vitro pull-down assay. To determine the functions of LGN in the central nervous system, we examined the expression and localization of LGN in mouse brain by immunoblotting and immunofluorescence microscopy. By immunoblotting, almost similar amounts of LGN were detected in the olfactory bulb, cerebral cortex, hippocampus, and cerebellum of the adult mouse brain, and the levels of the postnatal LGN expression in the whole brain were fairly constant. Immunofluorescence microscopy showed that LGN is localized in nuclei of the neurons in the olfactory bulb, cerebral cortex, and hippocampus, but in both nuclei and cytoplasm of Purkinje cells in the cerebellum. On the other hand, Galphai2 was distributed throughout the neuronal elements except for the nuclei. Thus, LGN and Galphai2 were colocalized in the cytoplasm of Purkinje cells, but not in other neurons examined. These results suggest that LGN may be involved not only in the Galphai2-mediated signaling but also in other signaling pathways.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...