Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 17(9): e0274033, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36067202

RESUMO

A novel risk locus at 4q32.2, located between the Nuclear Assembly Factor 1 (NAF1) and Follistatin Like 5 (FSTL5) genes, was associated with increased risk of developing colorectal cancer (CRC), with SNP rs17042479 being the most associated. However, the link between CRC development and the risk locus at 4q32.2 is unknown. We investigated the promoter activity of NAF1 and FSTL5 and analyzed the risk locus at 4q32.2 as gene regulatory region. Our results showed that the activity of the FSTL5 promoter was low compared to the NAF1 promoter. Analyses of the NAF1 promoter in conjunction with the region containing the risk locus at 4q32.2 showed that the region functions as gene regulatory region with repressor activity on NAF1 promoter activity. The SNP rs17042479(G) increased the repressor effect of the region. CRC patients' biopsies were genotyped for SNP rs17042479(A/G), and NAF1 expression profiles were examined. We found an association between SNP rs17042479(G), cancer stage and tumor location. Additionally, patients with SNP rs17042479(G) showed lower NAF1 expression in comparison to patients with SNP rs17042479(A) in tumor tissue and the NAF1 expression in tumor tissue was lower compared to healthy tissue. The results in the study imply that reduced NAF1 expression in the tumor contribute to a more aggressive phenotype. Furthermore, this study suggests that the SNP rs17042479(G) change the expression of NAF1 and thereby increases the risk of developing CRC.


Assuntos
Neoplasias Colorretais , Fatores de Transcrição NFI , Neoplasias Colorretais/genética , Genótipo , Humanos , Fatores de Transcrição NFI/genética , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas , Ribonucleoproteínas/genética
2.
FEBS Open Bio ; 11(6): 1638-1644, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33838073

RESUMO

Dysregulation of interleukin-33 (IL-33) has been implicated in the pathogenesis of several autoimmune and inflammatory diseases, but few studies have examined transcriptional regulation of the IL33 gene. In the intestines, gene regulation is controlled by a transcription factor network of which the intestinal-specific transcription factor CDX2 is a key component. In this study, we investigated whether CDX2 regulates IL33 mRNA expression. We examined IL33 mRNA expression in primary colonic epithelial cells from healthy humans and epithelial cell lines, revealing high expression levels in primary colonic and LS174T cells. Combining genomics data (ChIP-seq, RNA-seq) and IL33 promoter analyses in LS174T cells revealed intronic enhancer activity in the IL33 gene that is dependent on CDX2 expression. Western blotting and qRT-PCR confirmed that IL33 expression is upregulated in a CDX2 concentration-dependent manner, thereby providing the first evidence that CDX2 regulates the expression of IL33.


Assuntos
Fator de Transcrição CDX2/metabolismo , Células Epiteliais/metabolismo , Interleucina-33/genética , Intestinos/metabolismo , Fator de Transcrição CDX2/genética , Humanos , Interleucina-33/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células Tumorais Cultivadas
3.
BMC Cancer ; 20(1): 426, 2020 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-32408894

RESUMO

BACKGROUND: Colon cancer is one of the most commonly diagnosed types of cancer with surgical resection of the tumor being the primary choice of treatment. However, the surgical stress response induced during treatment may be related to a higher risk of recurrence. The aim of this study was to examine the effect of surgery on adhesion of cultured colon cancer cells with or without expression of the tumour suppressor CDX2. METHOD: We enrolled 30 patients undergoing elective, curatively intended laparoscopic surgery for colon cancer in this study. Blood samples were drawn 1 day prior to surgery and 24 h after surgery. The samples of pre- and postoperative serum was applied to wild type colon cancer LS174T cells and CDX2 inducible LS174T cells and adhesion was measured with Real-Time Cell-Analysis iCELLigence using electrical impedance as a readout to monitor changes in the cellular adhesion. RESULTS: Adhesion abilities of wild type LS174T cells seeded in postoperative serum was significantly increased compared to cells seeded in preoperative serum. When seeding the CDX2 inducible LS174T cells without CDX2 expression in pre- and postoperative serum, no significant difference in adhesion was found. However, when inducing CDX2 expression in these cells, the adhesion abilities in pre- and postoperative serum resembled those of the LS174T wild type cell line. CONCLUSIONS: We found that the adhesion of colon cancer cells was significantly increased in postoperative versus preoperative serum, and that CDX2 expression affected the adhesive ability of cancer cells. The results of this study may help to elucidate the pro-metastatic mechanisms in the perioperative phase and the role of CDX2 in colon cancer metastasis.


Assuntos
Biomarcadores Tumorais/metabolismo , Fator de Transcrição CDX2/metabolismo , Adesão Celular , Neoplasias do Colo/patologia , Laparoscopia/métodos , Assistência Perioperatória , Idoso , Movimento Celular , Neoplasias do Colo/sangue , Neoplasias do Colo/cirurgia , Feminino , Seguimentos , Humanos , Masculino , Prognóstico , Células Tumorais Cultivadas
4.
Int J Mol Sci ; 20(12)2019 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-31216773

RESUMO

The Hippo pathway is important for tissue homeostasis, regulation of organ size andgrowth in most tissues. The co-transcription factor yes-associated protein 1 (YAP1) serves as a maindownstream effector of the Hippo pathway and its dysregulation increases cancer development andblocks colonic tissue repair. Nevertheless, little is known about the transcriptional regulation ofYAP1 in intestinal cells. The aim of this study to identify gene control regions in the YAP1 gene andtranscription factors important for intestinal expression. Bioinformatic analysis of caudal typehomeobox 2 (CDX2) and hepatocyte nuclear factor 4 alpha (HNF4α) chromatin immunoprecipitatedDNA from differentiated Caco-2 cells revealed potential intragenic enhancers in the YAP1 gene.Transfection of luciferase-expressing YAP1 promoter-reporter constructs containing the potentialenhancer regions validated one potent enhancer of the YAP1 promoter activity in Caco-2 and T84cells. Two potential CDX2 and one HNF4α binding sites were identified in the enhancer by in silicotranscription factor binding site analysis and protein-DNA binding was confirmed in vitro usingelectrophoretic mobility shift assay. It was found by chromatin immunoprecipitation experimentsthat CDX2 and HNF4α bind to the YAP1 enhancer in Caco-2 cells. These results reveal a previouslyunknown enhancer of the YAP1 promoter activity in the YAP1 gene, with importance for highexpression levels in intestinal epithelial cells. Additionally, CDX2 and HNF4α binding areimportant for the YAP1 enhancer activity in intestinal epithelial cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Fator de Transcrição CDX2/metabolismo , Regulação da Expressão Gênica , Fator 4 Nuclear de Hepatócito/metabolismo , Intestinos , Fosfoproteínas/genética , Regiões Promotoras Genéticas , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Cromatina/genética , Cromatina/metabolismo , Elementos Facilitadores Genéticos , Humanos , Ligação Proteica , Fatores de Transcrição , Proteínas de Sinalização YAP
5.
Sci Rep ; 8(1): 11813, 2018 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-30087389

RESUMO

The type II membrane-anchored serine protease, matriptase, encoded by suppression of tumorgenicity-14 (ST14) regulates the integrity of the intestinal epithelial barrier in concert with its inhibitor, HAI-1 encoded by serine peptidase inhibitor, Kunitz type -1 (SPINT1). The balance of the protease/inhibitor gene expression ratio is vital in preventing the oncogenic potential of matriptase. The intestinal cell lineage is regulated by a transcriptional regulatory network where the tumor suppressor, Caudal homeobox 2 (CDX2) is considered to be an intestinal master transcription factor. In this study, we show that CDX2 has a dual function in regulating both ST14 and SPINT1, gene expression in intestinal cells. We find that CDX2 is not required for the basal ST14 and SPINT1 gene expression; however changes in CDX2 expression affects the ST14/SPINT1 mRNA ratio. Exploring CDX2 ChIP-seq data from intestinal cell lines, we identified genomic CDX2-enriched enhancer elements for both ST14 and SPINT1, which regulate their corresponding gene promoter activity. We show that CDX2 displays both repressive and enhancing regulatory abilities in a cell specific manner. Together, these data reveal new insight into transcriptional mechanisms controlling the intestinal matriptase/inhibitor balance.


Assuntos
Fator de Transcrição CDX2/metabolismo , Proteínas de Ciclo Celular/biossíntese , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Mucosa Intestinal/metabolismo , Proteínas Associadas aos Microtúbulos/biossíntese , Fosfoproteínas/biossíntese , Serina Endopeptidases/biossíntese , Fator de Transcrição CDX2/genética , Células CACO-2 , Proteínas de Ciclo Celular/genética , Células HeLa , Humanos , Mucosa Intestinal/citologia , Proteínas Associadas aos Microtúbulos/genética , Fosfoproteínas/genética , Serina Endopeptidases/genética
6.
PLoS One ; 13(7): e0200215, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29975781

RESUMO

Sequencing of primary colorectal tumors has identified a gene fusion in approximately 3% of colorectal cancer patients of the VTI1A and TCF7L2 genes, encoding a VTI1A-TCF4 fusion protein containing a truncated TCF4. As dysregulation of the Wnt signaling pathway is associated with colorectal cancer development and progression, the functional properties and transcriptional regulation of the VTI1A-TCF4 fusion protein may also play a role in these processes. Functional characteristics of the VTI1A-TCF4 fusion protein in Wnt signaling were analyzed in NCI-H508 and LS174T colon cancer cell lines. The NCI-H508 cell line, containing the VTI1A-TCF7L2 fusion gene, showed no active Wnt signaling, and overexpression of the VTI1A-TCF4 fusion protein in LS174T cells along with a Wnt signaling luciferase reporter plasmid showed inhibition of activity. The transcriptional regulation of the VTI1A-TCF4 fusion gene was investigated in LS174T cells where the activity of the VTI1A promoter was compared to that of the TCF7L2 promoter, and the transcription factor CDX2 was analyzed for gene regulatory activity of the VTI1A promoter through luciferase reporter gene assay using colon cancer cell lines as a model. Transfection of LS174T cells showed that the VTI1A promoter is highly active compared to the TCF7L2 promoter, and that CDX2 activates transcription of VTI1A. These results suggest that the VTI1A-TCF4 fusion protein is a dominant negative regulator of the Wnt signaling pathway, and that transcription of VTI1A is activated by CDX2.


Assuntos
Fator de Transcrição CDX2/genética , Neoplasias do Colo/genética , Proteínas de Fusão Oncogênica/genética , Proteínas Qb-SNARE/genética , Proteína 2 Semelhante ao Fator 7 de Transcrição/genética , Via de Sinalização Wnt , Sítios de Ligação , Fator de Transcrição CDX2/metabolismo , Linhagem Celular Tumoral , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Genes Reporter , Humanos , Intestinos/patologia , Proteínas de Fusão Oncogênica/metabolismo , Regiões Promotoras Genéticas , Proteínas Qb-SNARE/metabolismo , Proteína 2 Semelhante ao Fator 7 de Transcrição/metabolismo , beta Catenina/metabolismo
7.
Nat Commun ; 9(1): 1661, 2018 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-29695774

RESUMO

Inflammatory bowel disease (IBD) is a chronic intestinal disorder, with two main types: Crohn's disease (CD) and ulcerative colitis (UC), whose molecular pathology is not well understood. The majority of IBD-associated SNPs are located in non-coding regions and are hard to characterize since regulatory regions in IBD are not known. Here we profile transcription start sites (TSSs) and enhancers in the descending colon of 94 IBD patients and controls. IBD-upregulated promoters and enhancers are highly enriched for IBD-associated SNPs and are bound by the same transcription factors. IBD-specific TSSs are associated to genes with roles in both inflammatory cascades and gut epithelia while TSSs distinguishing UC and CD are associated to gut epithelia functions. We find that as few as 35 TSSs can distinguish active CD, UC, and controls with 85% accuracy in an independent cohort. Our data constitute a foundation for understanding the molecular pathology, gene regulation, and genetics of IBD.


Assuntos
Colite Ulcerativa/genética , Doença de Crohn/genética , Sequências Reguladoras de Ácido Nucleico/genética , Adulto , Biópsia , Estudos de Casos e Controles , Estudos de Coortes , Colite Ulcerativa/diagnóstico , Colite Ulcerativa/patologia , Colo/diagnóstico por imagem , Colo/patologia , Colonoscopia , Doença de Crohn/diagnóstico , Doença de Crohn/patologia , Feminino , Humanos , Mucosa Intestinal/diagnóstico por imagem , Mucosa Intestinal/patologia , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Análise de Sequência de RNA , Regulação para Cima
8.
Methods Mol Biol ; 1765: 57-77, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29589301

RESUMO

Several tumor suppressors possess gene regulatory activity. Here, we describe how promoter and promoter/enhancer reporter assays can be used to characterize a colorectal tumor suppressor proteins' gene regulatory activity of possible target genes. In the first part, a bioinformatic approach to identify relevant gene regulatory regions of potential target genes is presented. In the second part, it is demonstrated how to prepare and carry out the functional assay.We explain how to clone the bioinformatically identified gene regulatory regions into luciferase reporter plasmids by the use of the quick and efficient In-Fusion cloning method, and how to carry out transient transfections of Caco-2 colon cancer cells with the produced luciferase reporter plasmids using polyethyleneimine (PEI). A plan describing how to set up and carry out the luciferase expression assay is presented. The luciferase/ß-galactosidase (Dual Light) assay presented is a highly sensitive assay that can monitor small changes in the promoter/enhancer activity and includes an internal control monitoring transfection efficiency.


Assuntos
Bioensaio/métodos , Neoplasias Colorretais/genética , Regulação Neoplásica da Expressão Gênica , Regiões Promotoras Genéticas/genética , Proteínas Supressoras de Tumor/metabolismo , Bioensaio/instrumentação , Fator de Transcrição CDX2/metabolismo , Células CACO-2 , Clonagem Molecular/métodos , Biologia Computacional/métodos , Genes Reporter/genética , Humanos , Luciferases/química , Luciferases/genética , Glicoproteínas de Membrana/genética , Transcrição Gênica , Proteínas Supressoras de Tumor/genética
9.
J Cell Biochem ; 118(2): 298-307, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27333824

RESUMO

The expression of Caudal-related homeobox transcription factor 2 (CDX2) is impaired by tumor necrosis factor-α (TNF-α)-mediated activation of nuclear factor-κB (NF-κB) in ulcerative colitis (UC). Laminin subunit γ2 (LAMC2) is an epithelial basement membrane protein implicated in cell migration, proliferation, differentiation, as well as tumor invasion and intestinal inflammation, and its expression is enhanced by TNF-α in a NF-κB-dependent regulation of the recently identified LAMC2 enhancer. The aim was to determine whether CDX2 is involved in the basal regulation of LAMC2 in epithelial cells and to assess the influence of inflammation. Transcriptional regulation of LAMC2 was examined by reporter gene assays, overexpression, and shRNA-mediated knock-down of CDX2. CDX2-DNA interactions were assessed by chromatin immunoprecipitation on Caco-2 cells without or with TNF-α, as well as in purified colonic human epithelial cells. Immunohistochemical staining and quantitative reverse-transcription polymerase chain reaction analyses were used to measure the expression of CDX2 and LAMC2 in colonic biopsies from healthy controls and patients with UC. These data indicate that CDX2 directly regulates LAMC2 gene expression through interaction with elements in the LAMC2 promoter region. We further revealed an inverse effect of inflammation on CDX2 and LAMC2. The data presented provide a novel insight into how CDX2 is implicated in the transcriptional regulation of LAMC2 in intestinal epithelial cells, a function that is impaired during mucosal inflammation where a high level of TNF-α is present. J. Cell. Biochem. 118: 298-307, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Fator de Transcrição CDX2/biossíntese , Colite/metabolismo , Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Mucosa Intestinal/metabolismo , Laminina/metabolismo , Células CACO-2 , Colite/patologia , Colo , Células Epiteliais/patologia , Feminino , Humanos , Inflamação/metabolismo , Inflamação/patologia , Mucosa Intestinal/patologia , Masculino , Fator de Necrose Tumoral alfa/metabolismo
10.
DNA Res ; 21(6): 569-83, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24990076

RESUMO

The Caco-2 cell line is one of the most important in vitro models for enterocytes, and is used to study drug absorption and disease, including inflammatory bowel disease and cancer. In order to use the model optimally, it is necessary to map its functional entities. In this study, we have generated genome-wide maps of active transcription start sites (TSSs), and active enhancers in Caco-2 cells with or without tumour necrosis factor (TNF)-α stimulation to mimic an inflammatory state. We found 520 promoters that significantly changed their usage level upon TNF-α stimulation; of these, 52% are not annotated. A subset of these has the potential to confer change in protein function due to protein domain exclusion. Moreover, we locate 890 transcribed enhancer candidates, where ∼50% are changing in usage after TNF-α stimulation. These enhancers share motif enrichments with similarly responding gene promoters. As a case example, we characterize an enhancer regulating the laminin-5 γ2-chain (LAMC2) gene by nuclear factor (NF)-κB binding. This report is the first to present comprehensive TSS and enhancer maps over Caco-2 cells, and highlights many novel inflammation-specific promoters and enhancers.


Assuntos
Mapeamento Cromossômico , Elementos de Resposta/fisiologia , Iniciação da Transcrição Genética/efeitos dos fármacos , Iniciação da Transcrição Genética/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Células CACO-2 , Estudo de Associação Genômica Ampla , Humanos , Laminina/biossíntese , Laminina/genética , NF-kappa B/genética , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
11.
Proc Natl Acad Sci U S A ; 109(18): 6987-92, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22493273

RESUMO

Glycosphingolipids (GSLs) are of fundamental importance in the nervous system. However, the molecular details associated with GSL function are largely unknown, in part because of the complexity of GSL biosynthesis in vertebrates. In Drosophila, only one major GSL biosynthetic pathway exists, controlled by the glycosyltransferase Egghead (Egh). Here we discovered that loss of Egh causes overgrowth of peripheral nerves and attraction of immune cells to the nerves. This phenotype is reminiscent of the human disorder neurofibromatosis type 1, which is characterized by disfiguring nerve sheath tumors with mast cell infiltration, increased cancer risk, and learning deficits. Neurofibromatosis type 1 is due to a reduction of the tumor suppressor neurofibromin, a negative regulator of the small GTPase Ras. Enhanced Ras signaling promotes glial growth through activation of phosphatidylinositol 3-kinase (PI3K) and its downstream kinase Akt. We find that overgrowth of peripheral nerves in egh mutants is suppressed by down-regulation of the PI3K signaling pathway by expression of either dominant-negative PI3K, the tumor suppressor PTEN, or the transcription factor FOXO in the subperineurial glia. These results show that loss of the glycosyltransferase Egh affects membrane signaling and activation of PI3K signaling in glia of the peripheral nervous system, and suggest that glycosyltransferases may suppress proliferation.


Assuntos
Drosophila/metabolismo , Glucosilceramidas/metabolismo , Neurofibromatose 1/metabolismo , Animais , Modelos Animais de Doenças , Regulação para Baixo , Drosophila/genética , Drosophila/imunologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Genes de Insetos , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Microscopia Eletrônica de Transmissão , Modelos Neurológicos , Mutação , Neurofibromatose 1/genética , Neurofibromatose 1/imunologia , Neurofibromatose 1/patologia , Nervos Periféricos/imunologia , Nervos Periféricos/metabolismo , Nervos Periféricos/patologia , Fenótipo , Fosfatidilinositol 3-Quinases , Transdução de Sinais , Proteínas ras/metabolismo
12.
Genetics ; 188(4): 883-96, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21625003

RESUMO

The Drosophila melanogaster anterior-posterior axis is established during oogenesis by the localization of bicoid and oskar mRNAs to the anterior and posterior poles of the oocyte. Although genetic screens have identified some trans-acting factors required for the localization of these transcripts, other factors may have been missed because they also function at other stages of oogenesis. To circumvent this problem, we performed a screen for revertants and dominant suppressors of the bicaudal phenotype caused by expressing Miranda-GFP in the female germline. Miranda mislocalizes oskar mRNA/Staufen complexes to the oocyte anterior by coupling them to the bicoid localization pathway, resulting in the formation of an anterior abdomen in place of the head. In one class of revertants, Miranda still binds Staufen/oskar mRNA complexes, but does not localize to the anterior, identifying an anterior targeting domain at the N terminus of Miranda. This has an almost identical sequence to the N terminus of vertebrate RHAMM, which is also a large coiled-coil protein, suggesting that it may be a divergent Miranda ortholog. In addition, we recovered 30 dominant suppressors, including multiple alleles of the spectroplakin, short stop, a lethal complementation group that prevents oskar mRNA anchoring, and a female sterile complementation group that disrupts the anterior localization of bicoid mRNA in late oogenesis. One of the single allele suppressors proved to be a mutation in the actin nucleator, Cappuccino, revealing a previously unrecognized function of Cappuccino in pole plasm anchoring and the induction of actin filaments by Long Oskar protein.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Proteínas de Homeodomínio/metabolismo , Oócitos/metabolismo , RNA Mensageiro/metabolismo , Transativadores/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Drosophila/genética , Feminino , Genes Dominantes , Proteínas de Homeodomínio/genética , Masculino , Proteínas dos Microfilamentos/metabolismo , Dados de Sequência Molecular , Mutação/genética , Oogênese/genética , Alinhamento de Sequência , Transativadores/genética
13.
Dev Cell ; 13(4): 539-53, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17925229

RESUMO

Mutants in the actin nucleators Cappuccino and Spire disrupt the polarized microtubule network in the Drosophila oocyte that defines the anterior-posterior axis, suggesting that microtubule organization depends on actin. Here, we show that Cappuccino and Spire organize an isotropic mesh of actin filaments in the oocyte cytoplasm. capu and spire mutants lack this mesh, whereas overexpressed truncated Cappuccino stabilizes the mesh in the presence of Latrunculin A and partially rescues spire mutants. Spire overexpression cannot rescue capu mutants, but prevents actin mesh disassembly at stage 10B and blocks late cytoplasmic streaming. We also show that the actin mesh regulates microtubules indirectly, by inhibiting kinesin-dependent cytoplasmic flows. Thus, the Capu pathway controls alternative states of the oocyte cytoplasm: when active, it assembles an actin mesh that suppresses kinesin motility to maintain a polarized microtubule cytoskeleton. When inactive, unrestrained kinesin movement generates flows that wash microtubules to the cortex.


Assuntos
Actinas/fisiologia , Proteínas de Drosophila/metabolismo , Drosophila/ultraestrutura , Proteínas dos Microfilamentos/metabolismo , Microtúbulos/fisiologia , Oócitos/ultraestrutura , Animais , Citoplasma/fisiologia , Citoplasma/ultraestrutura , Drosophila/metabolismo , Proteínas de Drosophila/genética , Feminino , Cinesinas/metabolismo , Proteínas dos Microfilamentos/genética , Mutação , Oócitos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA