Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Biomed Sci ; 22: 102, 2015 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-26572230

RESUMO

BACKGROUND: Mutations in mitogen-activated protein kinase (MAPK) kinase 1 (MEK1) that occur during cell proliferation and tumor formation are well described. Information on the roles of MEK2 in these effects is still limited. We established a constitutive MEK2 transgenic zebrafish, Tg(krt14:MEK2S219D-GFP), to elucidate the role of MEK2 in skin tumor formation. RESULTS: We found that both constitutive MEK2 and MEK1 are able to phosphorylate the extracellular signal-regulated kinase 1 (ERK1) protein. Transient expression of constitutive MEK2 and MEK1 in the zebrafish epidermis induced papillary formation at 48 h post-fertilization, but no effects were observed due to the expression of MEK1, MEK2, or the dominant negative form of MEK2. The transgenic zebrafish, Tg(krt14:MEK2S219D-GFP), developed skin papillomas in the epidermis within 6 days post-fertilization (dpf). The phospho-ERK signal was detected in section of skin papillomas in an immunohistochemical experiment. Treatment with 50 µM of the MEK inhibitor, U0126, had significantly decreased the skin papilloma formation in Tg(krt14:MEK2S219D-GFP) zebrafish by 6 dpf. In vitro and in vivo proliferation assay in COS-1 cells and in Tg(krt14:MEK2S219D-GFP) transgenic fish show significantly increased cell number and Ki-67 signaling. CONCLUSION: Our data indicate that MEK2 is sufficient to induce epidermal papilloma formation through MAPK signaling in zebrafish, and this transgenic model can be used as a new platform for drug screening.


Assuntos
MAP Quinase Quinase 2/metabolismo , Papiloma/metabolismo , Neoplasias Cutâneas/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados/genética , Animais Geneticamente Modificados/metabolismo , Ativação Enzimática/genética , MAP Quinase Quinase 2/genética , Papiloma/genética , Neoplasias Cutâneas/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
2.
Fish Shellfish Immunol ; 38(1): 230-43, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24680831

RESUMO

Induction of interferons (IFNs) produces an innate immune response through activation of the JAK-STAT signaling pathway. Type I IFN signaling activates downstream gene expression through the IFN-stimulated gene factor 3 (ISGF3) complex, while type II IFN (IFN-γ) signaling is mediated through active STAT1 protein. The IFN target gene Mx is involved in the defense against viral infection. However, the mechanism by which Tetraodon (pufferfish) Mx is regulated by IFN signaling has not been identified. In this study, we describe the cloning and expression of Tetraodon STAT1, STAT2, and IFN regulatory factor 9 (IRF9). By combining constitutively-active STAT1 (STAT1-JH1) and STAT2 (STA2-JH1) fusion proteins with IRF9, we demonstrate that a constitutively-active ISGF3 complex increases the transcriptional activity of the Tetraodon Mx promoter via direct binding to two IFN-stimulated response element (ISRE) sites. In addition, a constitutively-active TnIRF9-S2C containing a fusion of the C-terminal region of STAT2 and IRF9 also activated the Mx promoter through binding to the ISRE sites. Furthermore, constitutively-active STAT1-JH1 elevates Mx promoter activity through two IFN gamma-activated sequence (GAS) elements. The Mx promoter is also activated by constitutively-active TnIRF9-S2C and STAT1-JH1 protein, as determined using an in vivo luciferase assay. We conclude that the Tetraodon Mx gene is activated via Type I (IFN-1) and Type II (IFN-γ) signaling. These results provide mechanistic insights into the role of IFN signaling in teleosts, and the in vivo luciferase assay may be suitable as a tool for studying induction and regulation by IFNs in teleost fish.


Assuntos
Regulação da Expressão Gênica/fisiologia , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/metabolismo , Proteínas de Resistência a Myxovirus/metabolismo , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT2/metabolismo , Tetraodontiformes/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Clonagem Molecular , DNA Complementar , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/genética , Dados de Sequência Molecular , Proteínas de Resistência a Myxovirus/genética , Filogenia , Regiões Promotoras Genéticas , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT2/genética , Transdução de Sinais
3.
Cell Mol Life Sci ; 70(13): 2367-81, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23361036

RESUMO

Protein activities controlled by receptor protein tyrosine phosphatases (RPTPs) play comparably important roles in transducing cell surface signals into the cytoplasm by protein tyrosine kinases. Previous studies showed that several RPTPs are involved in neuronal generation, migration, and axon guidance in Drosophila, and the vertebrate hippocampus, retina, and developing limbs. However, whether the protein tyrosine phosphatase type O (ptpro), one kind of RPTP, participates in regulating vertebrate brain development is largely unknown. We isolated the zebrafish ptpro gene and found that its transcripts are primarily expressed in the embryonic and adult central nervous system. Depletion of zebrafish embryonic Ptpro by antisense morpholino oligonucleotide knockdown resulted in prominent defects in the forebrain and cerebellum, and the injected larvae died on the 4th day post-fertilization (dpf). We further investigated the function of ptpro in cerebellar development and found that the expression of ephrin-A5b (efnA5b), a Fgf signaling induced cerebellum patterning factor, was decreased while the expression of dusp6, a negative-feedback gene of Fgf signaling in the midbrain-hindbrain boundary region, was notably induced in ptpro morphants. Further analyses demonstrated that cerebellar defects of ptpro morphants were partially rescued by inhibiting Fgf signaling. Moreover, Ptpro physically interacted with the Fgf receptor 1a (Fgfr1a) and dephosphorylated Fgfr1a in a dose-dependant manner. Therefore, our findings demonstrate that Ptpro activity is required for patterning the zebrafish embryonic brain. Specifically, Ptpro regulates cerebellar formation during zebrafish development through modulating Fgf signaling.


Assuntos
Cerebelo/embriologia , Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/embriologia , Animais , Padronização Corporal/genética , Diferenciação Celular , Sistema Nervoso Central/embriologia , Cerebelo/metabolismo , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário/genética , Técnicas de Silenciamento de Genes , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Transdução de Sinais , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
4.
Nucleic Acids Res ; 38(14): 4635-50, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20378713

RESUMO

In mammals, the Nogo family consists of Nogo-A, Nogo-B and Nogo-C. However, there are three Rtn-4/Nogo-related transcripts were identified in zebrafish. In addition to the common C-terminal region, the N-terminal regions of Rtn4-n/Nogo-C1, Rtn4-m/Nogo-C2 and Rtn4-l/Nogo-B, respectively, contain 9, 25 and 132 amino acid residues. In this study, we isolated the 5'-upstream region of each gene from a BAC clone and demonstrated that the putative promoter regions, P1-P3, are functional in cultured cells and zebrafish embryos. A transgenic zebrafish Tg(Nogo-B:GFP) line was generated using P1 promoter region to drive green fluorescent protein (GFP) expression through Tol2-mediated transgenesis. This line recapitulates the endogenous expression pattern of Rtn4-l/Nogo-B mRNA in the brain, brachial arches, eyes, muscle, liver and intestines. In contrast, GFP expressions by P2 and P3 promoters were localized to skeletal muscles of zebrafish embryos. Several GATA and E-box motifs are found in these promoter regions. Using morpholino knockdown experiments, GATA4 and GATA6 were involved in the control of P1 promoter activity in the liver and intestine, while Myf5 and MyoD for the control of P1 and P3 promoter activities in muscles. These data demonstrate that zebrafish Rtn4/Nogo transcripts might be generated by coupling mechanisms of alternative first exons and alternative promoter usage.


Assuntos
Proteínas da Mielina/genética , Regiões Promotoras Genéticas , Proteínas de Xenopus/genética , Peixe-Zebra/genética , Processamento Alternativo , Animais , Animais Geneticamente Modificados , Linhagem Celular , Embrião não Mamífero/metabolismo , Fatores de Transcrição GATA/metabolismo , Mucosa Intestinal/metabolismo , Fígado/metabolismo , Camundongos , Proteínas da Mielina/metabolismo , Proteínas Nogo , Proteínas de Xenopus/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra
5.
Fish Shellfish Immunol ; 28(5-6): 819-28, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20156562

RESUMO

In this paper, we report the cloning and characterization of the STAT6 gene from the pufferfish, Tetraodon nigroviridis. The TnSTAT6 gene is composed of 20 exons and 19 introns. The exon-intron organization of this gene is similar to that of HsSTAT6 except for the exons encoding the C-terminal transactivation domain. The full-length complementary (c)DNA of TnSTAT6 encodes a 794-amino acid protein that is 31% identical to human STAT6. We generated a constitutively active TnSTAT6-JH1 by fusing the kinase domain of carp JAK1 to the C-terminal end of TnSTAT6 and demonstrated that the fusion protein has specific DNA-binding ability and can activate a reporter construct carrying multiple copies of mammalian IL-4-response elements. Interestingly, TnSTAT6-JH1 associated with and phosphorylated TnSTAT6 on Tyr661. Mutation of this residue, Y661W, in TnSTAT6 abolished its association with TnSTAT6-JH1. This is consistent with the importance of the corresponding Tyr641 of HsSTAT6 in tyrosine phosphorylation and dimer formation. On the other hand, treatment of mammalian IL-4 did not induce tyrosine phosphorylation of wild-type TnSTAT6, suggesting that both the divergent N-terminal domain and coiled-coiled domain of TnSTAT6 may affect the interaction of TnSTAT6 with mammalian IL-4 receptor complexes.


Assuntos
Fator de Transcrição STAT6/genética , Fator de Transcrição STAT6/metabolismo , Tetraodontiformes/genética , Tetraodontiformes/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Clonagem Molecular , DNA Complementar/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Ordem dos Genes , Interleucina-4/farmacologia , Dados de Sequência Molecular , Fosforilação , Alinhamento de Sequência , Tirosina/metabolismo
6.
Fish Shellfish Immunol ; 28(5-6): 774-82, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20156563

RESUMO

In this study, we isolated and characterized both JAK and STAT genes from Artemia, Artemia franciscana. Although AfJAK showed only 19% identity (33% similarity) to the Drosophila Hop protein, AfJAK contained the characteristic JAK homology domain (JH domain) from JH1 to JH7. On the other hand, AfSTAT showed higher identity (30%) to Drosophila STAT (STAT92E). The low identities of AfJAK and AfSTAT to Drosophila Hop and STAT92E suggest that JAK and STAT proteins are unique in each different species of invertebrate. RT-PCR analysis showed that both AfJAK and AfSTAT transcripts were ubiquitously expressed in the embryo, which is similar to the expression patterns of Drosophila Hop and STAT92E mRNAs during development. In addition, we generated a constitutively active form of AfSTAT by fusing the JH1 domain of AfJAK to the C-terminal end of AfSTAT. This fusion protein, AfSTAT-HA-JH1, autophosphorylated on its tyrosine residue and was able to bind to specific DNA motifs including the STAT-binding motifs in the Drosophila Raf promoter. Both AfJAK and AfSTAT proteins elicited the transactivation potential toward the fly Raf promoter in Sf9 cells. However, tyrosine phosphorylation of AfSTAT was not detected, which is consistent with the cellular localization analysis that most AfSTAT proteins were in the cytoplasm. Our results demonstrate that both JAK and STAT are present in the genome of Artemia, which can serve as the basis for further investigations to explore the role of the JAK/STAT signal pathway in the development and immune response of brine shrimp.


Assuntos
Artemia/genética , Artemia/metabolismo , Regulação da Expressão Gênica , Janus Quinases/genética , Janus Quinases/metabolismo , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/metabolismo , Sequência de Aminoácidos , Animais , Artemia/citologia , Artemia/enzimologia , Células COS , Linhagem Celular , Chlorocebus aethiops , Clonagem Molecular , DNA/metabolismo , Perfilação da Expressão Gênica , Dados de Sequência Molecular , Filogenia , Regiões Promotoras Genéticas , Alinhamento de Sequência
7.
Cancer Res ; 67(5): 2089-97, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17332338

RESUMO

Osteopontin (OPN) and splice variants of CD44 (CD44(V)) have independently been identified as markers for tumor progression. In this study, we show that both OPN and CD44(V) are frequently overexpressed in human gastric cancer and that OPN-engaged CD44(V) ligation confers cells an increased survival mediated through integrin activation. First, we show that OPN treatment confers cells an increased resistance to UV-induced apoptosis. The OPN-mediated antiapoptosis is dependent on the expression of the variant exon 6 (V6)- or V7-containing CD44 as shown by overexpression of individual CD44(V) in gastric AZ521 cells that express no or very low level of endogenous CD44 and by knockdown of the constitutively expressed V6-containing CD44 isoforms in colon HT29 cells. Although OPN also interacts with RGD integrins, OPN-RGD sequence is dispensable for OPN-mediated antiapoptosis. OPN-induced antiapoptosis is mainly attributed to the engagement of CD44(V) isoforms and the relay of an inside-out signaling via Src activity, leading to robust integrin activation. Furthermore, OPN-elicited antiapoptosis was observed when cells were plated on fibronectin but not on poly-D-lysin, and preincubation of cells with anti-integrin beta(1) antibody to block integrin-extracellular matrix (ECM) interaction or ectopic expression of the dominant-negative forms of focal adhesion kinase to block ECM-derived signal abolished OPN-induced survival, suggesting that OPN-elicited antiapoptotic function is propagated from matrix transduced by integrin. Taken together, we showed that OPN-CD44(V) interaction promotes ECM-derived survival signal mediated through integrin activation, which may play an important role in the pathogenic development and progression of gastric cancer.


Assuntos
Adenocarcinoma/patologia , Neoplasias Gastrointestinais/patologia , Receptores de Hialuronatos/metabolismo , Integrinas/metabolismo , Osteopontina/fisiologia , Adenocarcinoma/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Adesão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Neoplasias Gastrointestinais/metabolismo , Células HT29 , Humanos , Osteopontina/metabolismo , Osteopontina/farmacologia , Isoformas de Proteínas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas , Raios Ultravioleta/efeitos adversos
8.
Dev Comp Immunol ; 32(7): 814-24, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18187191

RESUMO

In invertebrates, the JAK-STAT signaling pathway is involved in the anti-bacterial response and is part of an anti-viral response in Drosophila. In this study, we show that two STAT transcripts are generated by alternative splicing and encode two isoforms of Sf-STAT with different C-terminal ends. These two isoforms were produced and purified using the recombinant baculovirus technology. Both purified isoforms showed similar DNA-binding activity and displayed weak but significant transactivation potential toward a Drosophila promoter that contained a STAT-binding motif. No significant activation of the Sf-STAT protein in Sf9 cells was found by infection with baculovirus AcMNPV.


Assuntos
Expressão Gênica , Fatores de Transcrição STAT/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Núcleo Celular/metabolismo , DNA/metabolismo , Humanos , Dados de Sequência Molecular , Filogenia , Regiões Promotoras Genéticas/genética , Ligação Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/isolamento & purificação , Isoformas de Proteínas/metabolismo , Fatores de Transcrição STAT/química , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/isolamento & purificação , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Spodoptera , Ativação Transcricional/genética , Quinases raf/genética , Quinases raf/metabolismo
9.
FEBS Lett ; 581(22): 4265-71, 2007 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-17706649

RESUMO

In the present study, the zebrafish epo cDNA was cloned. The encoded protein displays 90%, 55% and 32% identity to the Epo from carp, fugu and human, respectively. Through RT-PCR, the expression of zepo mRNA was mainly in the heart and liver. In the COS-1 cell transfection experiments, the recombinant zEpo-HA protein was efficiently secreted into the culture medium as a glycoprotein and the carbohydrate moiety can be cleaved by the treatment of peptide-N-glycosidase F (PNGase F). Using the morpholino approach, we showed that zepo morphants displayed severe anemia leading to high mortality during development. Such an effect can be significantly rescued by zepo RNA. Furthermore, in the absence of functional zEpo, the expression of specific markers for adult globin genes, such as alphaA1- and betaA1-globin, but not the embryonic betae1-globin, was affected.


Assuntos
Eritropoetina/genética , Eritropoetina/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Clonagem Molecular , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Células Eritroides/metabolismo , Eritropoetina/química , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Hemoglobinas/biossíntese , Dados de Sequência Molecular , Especificidade de Órgãos , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Alinhamento de Sequência , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/química
10.
J Bone Miner Res ; 26(5): 1072-83, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21542008

RESUMO

Calcitonin (CT) is one of the hormones involved in vertebrate calcium regulation. It has been proposed to act as a hypocalcemic factor, but the regulatory pathways remain to be clarified. We investigated the CT/calcitonin gene-related peptide (CGRP) family in zebrafish and its potential involvement in calcium homeostasis. We identified the presence of four receptors: CTR, CRLR1, CRLR2, and CRLR3. From the phylogenetic analysis, together with the effect observed after CT and CGRP overexpression, we concluded that CTR appears to be a CT receptor and CRLR1 a CGRP receptor. The distribution of these two receptors shows a major presence in the central nervous system and in tissues involved in ionoregulation. Zebrafish embryos kept in high-Ca(2+)-concentration medium showed upregulation of CT and CTR expression and downregulation of the epithelial calcium channel (ECaC). Embryos injected with CT morpholino (CALC MO) incubated in high-Ca(2+) medium, showed downregulation of CTR together with upregulation on ECaC mRNA expression. In contrast, overexpression of CT cRNA induced the downregulation of ECaC mRNA synthesis, concomitant with the downregulation in the calcium content after 30 hours postfertilization. At 4 days postfertilization, CT cRNA injection induced upregulation of hypercalcemic factors, with subsequent increase in the calcium content. These results suggest that CT acts as a hypocalcemic factor in calcium regulation, probably through inhibition of ECaC synthesis.


Assuntos
Calcitonina/metabolismo , Cálcio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Homeostase/genética , Receptores da Calcitonina/metabolismo , Peixe-Zebra/metabolismo , Animais , Calcitonina/genética , Peptídeo Relacionado com Gene de Calcitonina/genética , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Meios de Cultura/farmacologia , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário/efeitos dos fármacos , Desenvolvimento Embrionário/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Proteínas de Fluorescência Verde/metabolismo , Homeostase/efeitos dos fármacos , Injeções , Oligonucleotídeos Antissenso/farmacologia , Especificidade de Órgãos/efeitos dos fármacos , Especificidade de Órgãos/genética , Filogenia , Plasmídeos/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores da Calcitonina/genética , Canais de Cátion TRPV , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
11.
PLoS One ; 6(10): e26461, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22028883

RESUMO

BACKGROUND: Mammalian M6A, a member of the proteolipid protein (PLP/DM20) family expressed in neurons, was first isolated by expression cloning with a monoclonal antibody. Overexpression of M6A was shown to induce filopodium formation in neuronal cells; however, the underlying mechanism of is largely unknown. Possibly due to gene duplication, there are two M6A paralogs, M6Aa and M6Ab, in the zebrafish genome. In the present study, we used the zebrafish as a model system to investigate the role of zebrafish M6Ab in filopodium formation in PC12 cells and neurite outgrowth in zebrafish embryos. METHODOLOGY/PRINCIPAL FINDINGS: We demonstrated that zebrafish M6Ab promoted extensive filopodium formation in NGF-treated PC12 cells, which is similar to the function of mammalian M6A. Phosphorylation at serine 263 of zebrafish M6Ab contributed to this induction. Transfection of the S263A mutant protein greatly reduced filopodium formation in PC12 cells. In zebrafish embryos, only S263D could induce neurite outgrowth. CONCLUSIONS/SIGNIFICANCE: Our results reveal that the phosphorylation status of zebrafish M6Ab at serine 263 is critical for its role in regulating filopodium formation and neurite outgrowth.


Assuntos
Embrião não Mamífero/citologia , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Neuritos/metabolismo , Pseudópodes/metabolismo , Serina/metabolismo , Proteínas de Peixe-Zebra/química , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Sequência de Aminoácidos , Animais , Células COS , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Chlorocebus aethiops , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/metabolismo , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/metabolismo , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 2/metabolismo , Glicoproteínas de Membrana/genética , Dados de Sequência Molecular , Fator de Crescimento Neural/farmacologia , Neuritos/efeitos dos fármacos , Células PC12 , Fosforilação/efeitos dos fármacos , Pseudópodes/efeitos dos fármacos , Ratos , Transdução de Sinais/efeitos dos fármacos , Proteínas de Peixe-Zebra/genética
12.
PLoS One ; 6(8): e23078, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21829695

RESUMO

BACKGROUND: The zona pellucida (ZP) domain is part of many extracellular proteins with diverse functions from structural components to receptors. The mammalian ß-tectorin is a protein of 336 amino acid residues containing a single ZP domain and a putative signal peptide at the N-terminus of the protein. It is 1 component of a gel-like structure called the tectorial membrane which is involved in transforming sound waves into neuronal signals and is important for normal auditory function. ß-Tectorin is specifically expressed in the mammalian and avian inner ear. METHODOLOGY/PRINCIPAL FINDINGS: We identified and cloned the gene encoding zebrafish ß-tectorin. Through whole-mount in situ hybridization, we demonstrated that ß-tectorin messenger RNA was expressed in the otic placode and specialized sensory patch of the inner ear during zebrafish embryonic stages. Morpholino knockdown of zebrafish ß-tectorin affected the position and number of otoliths in the ears of morphants. Finally, swimming behaviors of ß-tectorin morphants were abnormal since the development of the inner ear was compromised. CONCLUSIONS/SIGNIFICANCE: Our results reveal that zebrafish ß-tectorin is specifically expressed in the zebrafish inner ear, and is important for regulating the development of the zebrafish inner ear. Lack of zebrafish ß-tectorin caused severe defects in inner ear formation of otoliths and function.


Assuntos
Orelha Interna/embriologia , Proteínas da Matriz Extracelular/fisiologia , Peixe-Zebra/embriologia , Zona Pelúcida/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Primers do DNA , DNA Complementar , Proteínas da Matriz Extracelular/química , Proteínas da Matriz Extracelular/genética , Perfilação da Expressão Gênica , Humanos , Hibridização In Situ , Dados de Sequência Molecular , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Membrana Tectorial/metabolismo
13.
Dev Dyn ; 238(3): 746-54, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19235732

RESUMO

Human synuclein family consists of alpha-, beta-, and gamma-synucleins. Here, we cloned three genes, sncb, sncga and sncgb from zebrafish. They encode beta-, gamma1-, and gamma2-synucleins, respectively. The zSyn-beta, zSyn-gamma1, and zSyn-gamma2 proteins display 69%, 47%, and 50% identity to human beta-synuclein and gamma-synuclein, respectively. By reverse transcriptase-polymerase chain reaction, we demonstrated that sncb and sncga mRNA were abundant in brain and eye, while sncgb expression was moderate in brain, kidney, ovary and testis. The 1.8-kb 5'-upstream/promoter region of the sncga gene was sufficient to direct green fluorescent protein (GFP) expression in the central nervous system and cranial ganglions. A transgenic line, Tg(sncga:GFP), was generated and its GFP expression is similar to that of endogenous sncga mRNA. Moreover, this line also labels the habenular complex and the domain of GFP expression is larger in the left than in the right habenula. Thus, this line can be used to study sncga gene regulation and for left-right asymmetry study in zebrafish brain.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Habenula/metabolismo , Sinucleínas/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Envelhecimento/fisiologia , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Clonagem Molecular , Sequência Conservada , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Genes Reporter/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Habenula/embriologia , Habenula/crescimento & desenvolvimento , Humanos , Dados de Sequência Molecular , Filogenia , Regiões Promotoras Genéticas/genética , Ligação Proteica , Alinhamento de Sequência , Sinucleínas/química , Sinucleínas/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento , Proteínas de Peixe-Zebra/química , Proteínas de Peixe-Zebra/genética
14.
Biochem Biophys Res Commun ; 340(3): 767-75, 2006 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-16403448

RESUMO

We have previously identified a novel protein kinase, pk146, in the brain of Tetraodon. In the present study, we cloned the homologous protein kinase gene encoding a protein of 385 amino acid residues from zebrafish. The overall amino acid sequence and the kinase domain of zebrafish BSK146 shows 48% and 69% identity to that of rat sbk, a SH3-containing serine/threonine protein kinase. By whole-mount in situ hybridization and RT-PCR, the expression of bsk146 mRNA was mainly in the brain. To explore the in vivo function of BSK146 during zebrafish development, we used morpholino knockdown approach and found that BSK146 morphants displayed enlarged hindbrain ventricle and smaller eyes. Whole-mount in situ hybridization was further performed to analyze the brain defects in BSK146-MO-injected embryos. The expression of brain-specific markers, such as otx2, pax2.1, and krox20, was found normal in morphant embryos at 24hpf, while expression of pax2.1 exerted changes in midbrain-hindbrain boundary and hindbrain in morphant embryos at 48hpf. These data suggest that BSK146 may play an important role in later ventricle expansion in zebrafish brain development. Although the recombinant BSK146 protein produced in insect cells was active and could phosphorylate both histone H1 and histone 2B, the endogenous substrate of BSK146 in the embryonic brain of zebrafish is not clear at the present time and needs further investigation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Proteínas Quinases/biossíntese , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Sequência de Aminoácidos , Animais , Western Blotting , Encéfalo/embriologia , Encéfalo/metabolismo , Clonagem Molecular , Primers do DNA/química , DNA Complementar/metabolismo , Proteína 2 de Resposta de Crescimento Precoce/biossíntese , Éxons , Regulação da Expressão Gênica , Biblioteca Gênica , Genoma , Histonas/metabolismo , Hibridização In Situ , Insetos , Modelos Genéticos , Dados de Sequência Molecular , Mutação , Proteínas de Neoplasias/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fatores de Transcrição Otx/biossíntese , Fator de Transcrição PAX2/biossíntese , Fosforilação , Proteínas Serina-Treonina Quinases/biossíntese , Estrutura Terciária de Proteína , RNA/metabolismo , RNA Mensageiro/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Fatores de Tempo , Distribuição Tecidual , Peixe-Zebra , Proteínas de Peixe-Zebra/biossíntese
15.
Biochem Biophys Res Commun ; 344(1): 272-82, 2006 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-16616005

RESUMO

We expressed zebrafish p53 protein fused to GFP by a neuron-specific HuC promoter in zebrafish embryos. Instead of displaying neuronal expression patterns, p53-GFP was targeted to zebrafish YSL nuclei. This YSL targeting is p53 sequence-specific because GFP fusion proteins of p63 and p73 displayed neuronal-specific patterns. To dissect the underlying mechanisms, various constructs encoding a series of p53 mutant proteins under the control of different promoters were generated. Our results showed that expression of p53, in early zebrafish embryo, is preferentially targeted to the nuclei of YSL, which is mediated by importin. Similarly, this targeting is abrogated when p53 nuclear localization signal is disrupted. In addition, the transcriptional activity of p53 is required for this targeting. We further showed that fusion of pro-apoptotic BAD protein to p53-GFP led to apoptosis of YSL cells, and subsequent imperfect microtubule formation and abnormal blastomere movements.


Assuntos
Apoptose , Núcleo Celular/metabolismo , Transcrição Gênica , Proteína Supressora de Tumor p53/metabolismo , Saco Vitelino/metabolismo , Peixe-Zebra/embriologia , Animais , Bioensaio , Núcleo Celular/química , Citosol/química , Citosol/metabolismo , Proteínas ELAV/genética , Proteína Semelhante a ELAV 3 , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Carioferinas/metabolismo , Mutação , Neurônios/química , Neurônios/metabolismo , Sinais de Localização Nuclear/genética , Sinais de Localização Nuclear/metabolismo , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteína Supressora de Tumor p53/análise , Proteína Supressora de Tumor p53/genética , Saco Vitelino/química , Saco Vitelino/citologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/análise , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Proteína de Morte Celular Associada a bcl/análise , Proteína de Morte Celular Associada a bcl/genética , Proteína de Morte Celular Associada a bcl/metabolismo
16.
Genes Chromosomes Cancer ; 35(3): 219-31, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12353264

RESUMO

Gastric carcinoma (GC) is one of the most common malignancies worldwide and has a very poor prognosis. Genetic imbalances in 62 primary gastric adenocarcinomas of various histopathologic types and pathologic stages and six gastric cancer-derived cell lines were analyzed by comparative genomic hybridization, and the relationship of genomic abnormalities to clinical features in primary GC was evaluated at a genome-wide level. Eighty-four percent of the tumors and all six cell lines showed DNA copy number changes. The recurrent chromosomal abnormalities including gains at 15 regions and losses at 8 regions were identified. Statistical analyses revealed that gains at 17q24-qter (53%), 20q13-qter (48%), 1p32-p36 (42%), 22q12-qter (27%), 17p13-pter (24%), 16p13-pter (21%), 6p21-pter (19%), 20p12-pter (19%), 7p21-pter (18%), 3q28-qter (8%), and 13q13-q14 (8%), and losses at 18q12-qter (11%), 3p12 (8%), 3p25-pter (8%), 5q14-q23 (8%), and 9p21-p23 (5%), are associated with unique patient or tumor-related features. GCs of differing histopathologic features were shown to be associated with distinct patterns of genetic alterations, supporting the notion that they evolve through distinct genetic pathways. Metastatic tumors were also associated with specific genetic changes. These regions may harbor candidate genes involved in the pathogenesis of this malignancy.


Assuntos
Adenocarcinoma/genética , Aberrações Cromossômicas , Neoplasias Gástricas/genética , Adenocarcinoma/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Aneuploidia , Deleção Cromossômica , DNA de Neoplasias/genética , Feminino , Amplificação de Genes , Humanos , Hibridização in Situ Fluorescente/métodos , Interfase/genética , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Hibridização de Ácido Nucleico , Neoplasias Gástricas/patologia , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA