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.
Gene ; 767: 145148, 2021 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-32949698

RESUMO

Ischemic stroke is a common clinical cardiovascular disease and often accompanied by central nervous system injury. It often causes paralysis or loss of motor function after central nervous system injury and significantly reduces the patient's quality of life. At present, there is no effective treatment strategy for nerve damage caused by ischemic stroke. Therefore, it is urgently need to explore effective treatment targets. The protein expression of SOX5, VEGF and apoptosis related proteins were measured by western blot. The mRNA expression of SOX5 and VEGF were detected by RT-qPCR. The concentration of S100B and GFAP which are related to nerve damage were detected using ELISA assay. The transcriptional regulation of SOX5 on VEGF was detected using ChIP-PCR and dual luciferase reporter gene assays. The cell apoptosis was measured by TUNEL assay and cell viability was detected by CCK-8 assay. In our study, we found that the expression of SOX5 was significantly reduced when LPS induced apoptosis in PC-12 cells. Overexpression of SOX5 repaired LPS-induced apoptosis. SOX5 promotes VEGF expression as a transcription factor to activate the PI3K/AKT pathway. VEGF also repairs nerve injury and brain tissue injury caused by ischemic stroke. In conclusion, SOX5 transcription regulates the expression of VEGF to activate the PI3K/AKT pathway, which repaired nerve damage caused by ischemic stroke. Therefore, SOX5 could be a new targetto regulate VEGF which can repair nerve injury induced by ischemic stroke.


Assuntos
AVC Isquêmico/tratamento farmacológico , AVC Isquêmico/metabolismo , Fatores de Transcrição SOXD/metabolismo , Animais , Apoptose/fisiologia , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Morte Celular , Proliferação de Células , Regulação da Expressão Gênica/genética , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Células PC12 , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Fatores de Transcrição SOXD/genética , Fatores de Transcrição SOXD/fisiologia , Transdução de Sinais/genética , Acidente Vascular Cerebral/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fatores de Crescimento do Endotélio Vascular/metabolismo
2.
Mol Med Rep ; 22(2): 948-956, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32468049

RESUMO

Dilated cardiomyopathy (DCM) is a disease that can lead to heart expansion and severe heart failure, but the specific pathogenesis remains unclear. Sox5 is a member of the Sox family with a key role in cardiac function. However, the role of Sox5 in DCM remains unclear. In the present study, wild­type mice were intraperitoneally injected with doxorubicin (Dox) to induce DCM, and heart specimens from human patients with DCM were used to investigate the preliminary role of Sox5 in DCM. The present study demonstrated that, compared with control human hearts, the hearts of patients with DCM exhibited high expression levels of Sox5 and activation of the wnt/ß­catenin pathway. This result was consistent with Dox­induced DCM in mice. Furthermore, in Dox­treated mice, apoptosis was activated during the development of DCM. Inflammation and collagen deposition also increased in DCM mice. The results of the present study indicate that Sox5 may be associated with the development of DCM. Sox5 may be a novel potential factor that regulates DCM.


Assuntos
Cardiomiopatia Dilatada/metabolismo , Fatores de Transcrição SOXD/biossíntese , Fatores de Transcrição SOXD/fisiologia , Idoso , Animais , Apoptose , Cardiomiopatia Dilatada/induzido quimicamente , Colágeno/metabolismo , Modelos Animais de Doenças , Doxorrubicina/efeitos adversos , Feminino , Fibrose/metabolismo , Humanos , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Miocárdio/metabolismo , Miocárdio/patologia , Proteína Wnt1/metabolismo , beta Catenina/metabolismo
3.
Biomed Pharmacother ; 109: 2128-2135, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30551470

RESUMO

Altered expression of microRNAs (miRNAs) was involved in prostate cancer progression. However, how miRNAs contributed to prostate cancer development remained unknown. Here, we reported that miR-139-5p levels were decreased in prostate cancer tumor tissues and prostate cancer cell lines. Transfection of miR-139-5p mimics reduced cell proliferation and migration ability of prostate cancer cells. Western blotting and RT-qPCR showed that elevation of miR-139-5p greatly inhibited SOX5 expression in prostate cancer cells. Through regulation of SOX5, enhanced expression of miR-139-5p downregulated TWIST, decreased N-cadherin and Vimentin expression, suggesting inhibition of epithelial-mesenchymal transition (EMT) process. The dual luciferase assay validated that SOX5 was a direct target of miR-139-5p. Additionally, a significant negative correlation between SOX5 mRNA levels and miR-139-5p levels were detected in prostate cancer tumor tissues. Our study indicated that miR-139-5p functioned as a tumor suppressor in prostate cancer cells by regulation of SOX5, and it might be a promising target for prostate cancer patients.


Assuntos
Progressão da Doença , Regulação para Baixo/fisiologia , MicroRNAs/biossíntese , Neoplasias da Próstata/metabolismo , Fatores de Transcrição SOXD/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Masculino , MicroRNAs/genética , Neoplasias da Próstata/patologia
4.
Cell ; 175(2): 372-386.e17, 2018 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-30270042

RESUMO

Intestinal mesenchymal cells play essential roles in epithelial homeostasis, matrix remodeling, immunity, and inflammation. But the extent of heterogeneity within the colonic mesenchyme in these processes remains unknown. Using unbiased single-cell profiling of over 16,500 colonic mesenchymal cells, we reveal four subsets of fibroblasts expressing divergent transcriptional regulators and functional pathways, in addition to pericytes and myofibroblasts. We identified a niche population located in proximity to epithelial crypts expressing SOX6, F3 (CD142), and WNT genes essential for colonic epithelial stem cell function. In colitis, we observed dysregulation of this niche and emergence of an activated mesenchymal population. This subset expressed TNF superfamily member 14 (TNFSF14), fibroblastic reticular cell-associated genes, IL-33, and Lysyl oxidases. Further, it induced factors that impaired epithelial proliferation and maturation and contributed to oxidative stress and disease severity in vivo. Our work defines how the colonic mesenchyme remodels to fuel inflammation and barrier dysfunction in IBD.


Assuntos
Doenças Inflamatórias Intestinais/fisiopatologia , Mesoderma/fisiologia , Animais , Proliferação de Células , Colite/genética , Colite/fisiopatologia , Colo/fisiologia , Células Epiteliais/metabolismo , Fibroblastos/fisiologia , Heterogeneidade Genética , Homeostase , Humanos , Inflamação , Mucosa Intestinal/imunologia , Mucosa Intestinal/fisiologia , Intestinos/imunologia , Intestinos/fisiologia , Células-Tronco Mesenquimais/fisiologia , Mesoderma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Miofibroblastos , Pericitos , Células RAW 264.7 , Fatores de Transcrição SOXD/fisiologia , Análise de Célula Única/métodos , Tromboplastina/fisiologia , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Via de Sinalização Wnt/fisiologia
5.
Cancer Res ; 77(18): 4985-4997, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28687615

RESUMO

Molecular mechanisms preventing self-renewing brain stem cells from oncogenic transformation are poorly defined. We show that the expression levels of SOX5, SOX6, and SOX21 (SOX5/6/21) transcription factors increase in stem cells of the subventricular zone (SVZ) upon oncogenic stress, whereas their expression in human glioma decreases during malignant progression. Elevated levels of SOX5/6/21 promoted SVZ cells to exit the cell cycle, whereas genetic ablation of SOX5/6/21 dramatically increased the capacity of these cells to form glioma-like tumors in an oncogene-driven mouse brain tumor model. Loss-of-function experiments revealed that SOX5/6/21 prevent detrimental hyperproliferation of oncogene expressing SVZ cells by facilitating an antiproliferative expression profile. Consistently, restoring high levels of SOX5/6/21 in human primary glioblastoma cells enabled expression of CDK inhibitors and decreased p53 protein turnover, which blocked their tumorigenic capacity through cellular senescence and apoptosis. Altogether, these results provide evidence that SOX5/6/21 play a central role in driving a tumor suppressor response in brain stem cells upon oncogenic insult. Cancer Res; 77(18); 4985-97. ©2017 AACR.


Assuntos
Neoplasias Encefálicas/patologia , Transformação Celular Neoplásica/patologia , Glioma/patologia , Células-Tronco Neoplásicas/patologia , Fatores de Transcrição SOXB2/metabolismo , Fatores de Transcrição SOXD/fisiologia , Animais , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Senescência Celular , Feminino , Glioma/genética , Glioma/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Células-Tronco Neoplásicas/metabolismo , Oncogenes , Fatores de Transcrição SOXB2/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
6.
J BUON ; 22(1): 258-264, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28365963

RESUMO

PURPOSE: SOX5 plays important roles in various kinds of cancers. However, the expression and roles of SOX5 in osteosarcoma (OS) have not been investigated well. In the present study we aimed to investigate the mechanism of SOX5 in OS. METHODS: OS and adjacent non-cancerous specimens were obtained from patients with OS. PCR was applied to detect SOX5 mRNA. Then human OS cell lines (U2OS, SoSP-M, SoSP-9607, and MG-63) and one immortalized normal osteoblast hFOB1.19 were investigated. SOX5 knocking with shRNA in U2OS and SOX5 upregulation with recombinant plasmid in MG-63 were applied. Real-time cell monitoring system and invasion assay were used, and Western blot assay was performed to detect the protein level of E-cadherin, N-cadherin, Vimentin and Snail, where Glyceraldehyde3- phosphate dehydrogenase (GAPDH) was presented as control. P<0.05 was considered as statistically significant. RESULTS: Significant upregulation of SOX5 in OS tissues and cell lines was identified. The gain- and loss-of-function studies suggested that OS cell migration and invasion were promoted significantly by SOX5. Additionally, SOX5 promoted epithelial-mesenchymal transition (EMT) by regulation of Snail. CONCLUSION: SOX5 is a novel regulator of EMT in OS, and is a potential target for OS.


Assuntos
Neoplasias Ósseas/patologia , Transição Epitelial-Mesenquimal , Osteossarcoma/patologia , Fatores de Transcrição SOXD/fisiologia , Fatores de Transcrição da Família Snail/fisiologia , Neoplasias Ósseas/mortalidade , Linhagem Celular Tumoral , Movimento Celular , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Humanos , Invasividade Neoplásica , Osteossarcoma/mortalidade , Proteínas Proto-Oncogênicas c-raf/fisiologia
7.
Oncogene ; 35(13): 1692-702, 2016 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-26119940

RESUMO

Sex-determining region Y box 6 (SOX6) has been described as a tumor-suppressor gene in several cancers. Our previous work has suggested that SOX6 upregulated p21(Waf1/Cip1)(p21) expression in a p53-dependent manner; however, the underlying mechanism has remained elusive. In this study, we confirmed that SOX6 can suppress cell proliferation in vitro and in vivo by stabilizing p53 protein and subsequently upregulating p21. Co-immunoprecipitation and immunocytofluorescence assays demonstrated that SOX6 can promote formation of the p14ARF-HDM2-p53 ternary complex by promoting translocation of p14ARF (p14 alternate reading frame tumor suppressor) to the nucleoplasm, thereby inhibiting HDM2-mediated p53 nuclear export and degradation. Chromatin immunoprecipitation combined with PCR assay proved that SOX6 can bind to a potential binding site in the regulatory region of the c-Myc gene. Furthermore, we confirmed that SOX6 can downregulate the expression of c-Myc, as well as its direct target gene nucleophosmin 1 (NPM1), and that the SOX6-induced downregulation of NPM1 is linked to translocation of p14ARF to the nucleoplasm. Finally, we showed that the highly conserved high-mobility group (HMG) domain of SOX6 is required for SOX6-mediated p53 stabilization and tumor inhibitory activity. Collectively, these results reveal a new mechanism of SOX6-mediated tumor suppression involving p21 upregulation via the p14ARF-HDM2-p53 axis in an HMG domain-dependent manner.


Assuntos
Transformação Celular Neoplásica/genética , Proteínas Proto-Oncogênicas c-mdm2/fisiologia , Fatores de Transcrição SOXD/fisiologia , Proteína Supressora de Tumor p14ARF/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Proteína Supressora de Tumor p53/farmacocinética , Animais , Proliferação de Células/genética , Feminino , Genes Supressores de Tumor/fisiologia , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Nucleofosmina , Estrutura Terciária de Proteína/genética , Fatores de Transcrição SOXD/química , Fatores de Transcrição SOXD/genética , Transdução de Sinais/fisiologia , Células Tumorais Cultivadas
8.
J Pharmacol Sci ; 122(4): 318-25, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23883486

RESUMO

Pre-chondrocytes undergo cellular differentiation stages during chondrogenesis under the influence by different transcription factors such as sry-type high mobility group box-9 (Sox9) and runt-related transcription factor-2 (Runx2). We have shown upregulation by parathyroid hormone (PTH) of the clock gene Period-1 (Per1) through the cAMP/protein kinase A signaling pathway in pre-chondrocytic ATDC5 cells. Here, we investigated the role of Per1 in the suppression of chondrogenic differentiation by PTH. In ATDC5 cells exposed to 10 nM PTH, a drastic but transient increase in Per1 expression was seen only 1 h after addition together with a prolonged decrease in Sox6 levels. However, no significant changes were induced in Sox5 and Runx2 levels in cells exposed to PTH. In stable Per1 transfectants, a significant decrease in Sox6 levels was seen, with no significant changes in Sox5 and Sox9 levels, in addition to the inhibition of gene transactivation by Sox9 allies. Knockdown of Per1 by siRNA significantly increased the Sox6 and type II collagen levels in cells cultured for 24 - 60 h. These results suggest that Per1 plays a role in the suppressed chondrocytic differentiation by PTH through a mechanism relevant to negative regulation of transactivation of the Sox6 gene during chondrogenesis.


Assuntos
Diferenciação Celular/genética , Condrócitos/citologia , Condrogênese/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/fisiologia , Fatores de Transcrição SOXD/genética , Fatores de Transcrição SOXD/fisiologia , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Condrogênese/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Camundongos , Hormônio Paratireóideo/farmacologia , Proteínas Circadianas Period/metabolismo , RNA Interferente Pequeno , Fatores de Transcrição SOXD/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Ativação Transcricional/genética
9.
Osteoarthritis Cartilage ; 19(10): 1246-53, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21843649

RESUMO

OBJECTIVE: This study aims to investigate the regulation of expression of Cartilage oligomeric matrix protein (COMP), which is predominately expressed by chondrocytes and functions to organize the extracellular matrix. Mutations in COMP cause two skeletal dysplasias: pseudoachondroplasia and multiple epiphyseal dysplasia. The mechanism controlling COMP expression during chondrocyte differentiation is still poorly understood. DESIGN: Primary human bone marrow-derived stem cells were induced to differentiate into chondrocyte by pellet cultures. We then compared the temporal expression of COMP with the well-characterized cartilage-specific Type II collagen (Col2a1), and their response to transforming growth factor (TGF)ß and Sox trio (Sox5, 6, and 9) stimulation. RESULTS: COMP and Col2a1 expression are differentially regulated by three distinct mechanisms. First, upregulation of COMP mRNA precedes Col2a1 by several days during chondrogenesis. Second, COMP expression is independent of high cell density but requires TGF-ß1. Induction of COMP mRNA by TGF-ß1 is detected within 2h in the absence of protein synthesis and is blocked by specific inhibitors of the TGFß signaling pathway; and therefore, COMP is a primary TFGß-response gene. Lastly, while Col2a1 expression is intimately controlled by the Sox trio, overexpression of Sox trio fails to activate the COMP promoter. CONCLUSION: COMP and Col2a1 expression are regulated differently during chondrogenesis. COMP is a primary response gene of TGFß and its fast induction during chondrogenesis suggests that COMP is suitable for rapidly accessing the chondrogenic potential of stem cells.


Assuntos
Células da Medula Óssea/citologia , Condrogênese/fisiologia , Colágeno Tipo II/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Glicoproteínas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Fator de Crescimento Transformador beta1/fisiologia , Células da Medula Óssea/metabolismo , Proteína de Matriz Oligomérica de Cartilagem , Regulação da Expressão Gênica , Humanos , Proteínas Matrilinas , Células-Tronco Mesenquimais/citologia , Fatores de Transcrição SOX9/fisiologia , Fatores de Transcrição SOXD/fisiologia , Transdução de Sinais , Regulação para Cima
10.
Arthritis Rheum ; 62(10): 2864-75, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20533289

RESUMO

OBJECTIVE: We previously observed the association of the co-occurrence of the HLA-DRB1 shared epitope (SE) and RANKL single-nucleotide polymorphisms (SNPs) with younger age at the onset of rheumatoid arthritis (RA) in 182 rheumatoid factor (RF)-positive European American patients with early-onset RA. The aim of this study was to fine-map the 48-kb RANKL region in the extended cohort of 210 European American RF-positive patients with early RA, to seek replication of RA-associated SNPs in additional RA cohorts of 501 European Americans and 298 African Americans, and to explore the functional consequences of RA-associated SNPs. METHODS: SNP genotyping was conducted using pyrosequencing or TaqMan polymerase chain reaction (PCR) assays. Associations of rs7984870 with RANKL expression in plasma, peripheral blood mononuclear cells, and isolated T cells were quantified using enzyme-linked immunosorbent assay and reverse transcription-PCR. Site-directed mutagenesis of rs7984870 within the 2-kb RANKL promoter was performed to drive the luciferase reporter gene in osteoblast and stromal cell lines. Interaction of DNA and protein was determined by electrophoretic mobility shift assay. RESULTS: A single promoter SNP, rs7984870, was consistently significantly associated with earlier age at the onset of RA in 3 independent seropositive (RF or anti-cyclic citrullinated peptide antibody) RA cohorts but not in seronegative RA patients. The C risk allele of rs7984870 conferred 2-fold higher plasma RANKL levels in RF-positive patients with RA, significantly elevated RANKL messenger RNA expression in activated normal T cells, and increased promoter activity after stimulation in vitro via differential binding to the transcription factor SOX5. CONCLUSION: The RANKL promoter allele that increased transcription levels upon stimulation might promote interaction between activated T cells and dendritic cells, predisposing to a younger age at the onset of RA in seropositive European American and African American patients.


Assuntos
Artrite Reumatoide/genética , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas/genética , Ligante RANK/genética , Adulto , Negro ou Afro-Americano/genética , Idade de Início , Artrite Reumatoide/etnologia , Feminino , Predisposição Genética para Doença/etnologia , Predisposição Genética para Doença/genética , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Ligante RANK/sangue , RNA Mensageiro/metabolismo , Fatores de Transcrição SOXD/fisiologia , População Branca/genética
11.
Oncogene ; 28(12): 1537-48, 2009 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-19219070

RESUMO

SOX5 is a member of the high-mobility group superfamily of architectural non-histone proteins involved in gene regulation and maintenance of chromatin structure in a wide variety of developmental processes. Sox5 was identified as a brain tumor locus in a retroviral insertional mutagenesis screen of platelet-derived growth factor B (PDGFB)-induced mouse gliomas. Here we have investigated the role of Sox5 in PDGFB-induced gliomagenesis in mice. We show that Sox5 can suppress PDGFB-induced glioma development predominantly upon Ink4a-loss. In human glioma cell lines and tissues, we found very low levels of SOX5 compared with normal brain. Overexpression of Sox5 in human glioma cells led to a reduction in clone formation and inhibition of proliferation. Combined expression of Sox5 and PDGFB in primary brain cell cultures caused decreased proliferation and an increased number of senescent cells in the Ink4a-/- cells only. Protein analyses showed a reduction in the amount and activation of Akt and increased levels of p27(Kip1) upon Sox5 expression that was dominant to PDGFB signaling and specific to Ink4a-/- cells. Upon inhibition of p27(Kip1), the effects of Sox5 on proliferation and senescence could be reversed. Our data suggest a novel pathway, where Sox5 may suppress the oncogenic effects of PDGFB signaling during glioma development by regulating p27(Kip1) in a p19(Arf)-dependent manner, leading to acute cellular senescence.


Assuntos
Senescência Celular , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Glioma/prevenção & controle , Proteínas Proto-Oncogênicas c-sis/fisiologia , Fatores de Transcrição SOXD/fisiologia , Animais , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Glioma/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Camundongos , Proteínas Proto-Oncogênicas c-sis/antagonistas & inibidores , Células Tumorais Cultivadas
12.
Int J Cancer ; 124(10): 2323-32, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19173284

RESUMO

Identification of genomic alterations associated with the progression of prostate cancer may facilitate the better understanding of the development of this highly variable disease. Matched normal, premalignant high-grade prostatic intraepithelial neoplasia and invasive prostate carcinoma cells were procured by laser capture microdissection (LCM) from human radical prostatectomy specimens. From these cells, comparative DNA fingerprints were generated by a modified PCR-based technique called scanning of microdissected archival lesion (SMAL)-PCR. Recurrent polymorphic fingerprint fragments were used in tagging altered chromosomal regions. Altered regions were found at cytobands 1p31.3, 1q44, 2p23.1, 3p26.3, 3q22.3, 4q22.3, 4q35.2, 5q23.2, 8q22.3, 8q24.13, 9q21.3, 9q22.32, 10q11.21, 11p13, 12p12.1, 13q12.1, 16q12.2 and 18q21.31. Candidate genes in the surrounding area that may possibly harbor mutations that change normal prostatic cells to progress into their tumor stages were proposed. Of these fragments, a 420 bp alteration, absent in all 26 normal samples screened, was observed in 2 tumors. This fragment was cloned, sequenced and localized to chromosome 12p12.1. Within this region, candidate gene sex determining region Y-box 5 (SOX5) was proposed. Further studies of SOX5 in cell lines, xenografts and human prostate specimens, at both the RNA and protein levels, found overexpression of the gene in tumors. This overexpression was then subsequently found by fluorescent in situ hybridization to be caused by amplification of the region. In conclusion, our results suggest LCM coupled with SMAL-PCR DNA fingerprinting is a useful method for the screening and identification of chromosomal regions and genes associated with cancer development. Further, overexpression of SOX5 is associated with prostate tumor progression and early development of distant metastasis.


Assuntos
Mapeamento Cromossômico , Impressões Digitais de DNA , Neoplasias da Próstata/genética , Fatores de Transcrição SOXD/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Sequência de Bases , Linhagem Celular Tumoral , Primers do DNA , Progressão da Doença , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Fatores de Transcrição SOXD/genética , Análise Serial de Tecidos
13.
Proc Natl Acad Sci U S A ; 105(41): 16021-6, 2008 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-18840685

RESUMO

Neocortical projection neurons exhibit layer-specific molecular profiles and axonal connections. Here we show that the molecular identities of early-born subplate and deep-layer neurons are not acquired solely during generation or shortly thereafter but undergo progressive postmitotic refinement mediated by SOX5. Fezf2 and Bcl11b, transiently expressed in all subtypes of newly postmigratory early-born neurons, are subsequently downregulated in layer 6 and subplate neurons, thereby establishing their layer 5-enriched postnatal patterns. In Sox5-null mice, this downregulation is disrupted, and layer 6 and subplate neurons maintain an immature differentiation state, abnormally expressing these genes postnatally. Consistent with this disruption, SOX5 binds and represses a conserved enhancer near Fezf2. The Sox5-null neocortex exhibits failed preplate partition and laminar inversion of early-born neurons, loss of layer 5 subcerebral axons, and misrouting of subplate and layer 6 corticothalamic axons to the hypothalamus. Thus, SOX5 postmitotically regulates the migration, postmigratory differentiation, and subcortical projections of subplate and deep-layer neurons.


Assuntos
Diferenciação Celular , Movimento Celular , Extensões da Superfície Celular , Neocórtex/citologia , Neurônios/citologia , Fatores de Transcrição SOXD/fisiologia , Animais , Moléculas de Adesão Celular/genética , Proteínas de Ligação a DNA/genética , Regulação para Baixo/genética , Camundongos , Camundongos Knockout , Mitose , Proteínas Repressoras/genética , Proteínas Supressoras de Tumor/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA