Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Cell Biol Int ; 45(6): 1296-1305, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33739578

RESUMO

The prognosis of advanced colorectal cancer (CRC) is currently still very poor, which suggests that the biological mechanisms of CRC oncogenesis are not fully understood. This study was conducted to explore the regulatory effect of SOX-17 on the expression of microRNA (miR)-302b-3p, and the involvement of SOX-17 in the invasion and apoptosis of CRC cells. The expression of SOX-17 and miR-302a,b,c,d-3p in colorectal cancer and normal colon epithelial cell lines was measured by real-time polymerase chain reaction and/or western blot. The regulatory effects of SOX-17 on miR-302b-3p gene in HT29 and LoVo cells were tested using the ChiP assay. The biological activities of SOX-17 and miR-302b-3p were evaluated by invasion and apoptosis assay. Results showed that transfection of SOX-17 small interfering RNA (siSOX-17) significantly increased, whereas transfection of SOX-17 overexpression vector (oeSOX-17) significantly decreased, miR-302b expression in HT29 and LoVo cells. Cotransfection of oeSOX-17 and miR-302b-3p inhibitor (INmiR-302b) significantly blocked the effects of SOX-17 in HT29 and LoVo cells. ChIP experiments showed that SOX-17 bonded to the miR-302b-3p promoter in HT29 and LoVo cells. Transfection of oeSOX-17 and miR-302b-3p mimics (MImiR-302b) significantly decreased, whereas transfection of siSOX-17 and INmiR-302b significantly increased, the invasion of HT29 and LoVo cells. In contrast, transfection of oeSOX-17 and MImiR-302b significantly increased, while transfection of siSOX-17 and INmiR-302b significantly decreased, apoptosis in HT29 and LoVo cells. Cotransfection of oeSOX-17 and INmiR-302b significantly blocked the effects of oeSOX-17 on cell invasion and apoptosis in HT29 and LoVo cells. These results suggested that SOX-17 can bind to the promoter of miR-302b-3p gene to regulate its expression, while both SOX-17 and miR-302b regulate the invasion and apoptosis in colorectal cancer cells.


Assuntos
Neoplasias Colorretais/metabolismo , MicroRNAs/fisiologia , Fatores de Transcrição SOXF/fisiologia , Apoptose , Linhagem Celular Tumoral , Movimento Celular , Regulação Neoplásica da Expressão Gênica , Humanos
2.
Surg Today ; 51(3): 340-349, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32754843

RESUMO

Organ liver transplantation and hepatocyte transplantation are not performed to their full potential because of donor shortage, which could be resolved by identifying new donor sources for the development of hepatocyte-like cells (HLCs). HLCs have been differentiated from some stem cell sources as alternative primary hepatocytes throughout the world; however, the currently available techniques cannot differentiate HLCs to the level of normal adult primary hepatocytes. The outstanding questions are as follows: which stem cells are the best cell sources? which protocol is the best way to differentiate them into HLCs? what is the definition of differentiated HLCs? how can we enforce the function of HLCs? what is the difference between HLCs and primary hepatocytes? what are the problems with HLC transplantation? This review summarizes the current status of HLCs, focusing on stem cell sources, the differentiation protocol for HLCs, the general characterization of HLCs, the generation of more functional HLCs, comparison with primary hepatocytes, and HLCs in cell-transplantation-based liver regeneration.


Assuntos
Diferenciação Celular , Terapia Baseada em Transplante de Células e Tecidos/métodos , Técnicas Citológicas/métodos , Hepatócitos/transplante , Hepatopatias/terapia , Células-Tronco/fisiologia , Proteínas Morfogenéticas Ósseas/fisiologia , Diferenciação Celular/genética , Células Cultivadas , Técnicas de Cocultura , Fatores de Crescimento de Fibroblastos/fisiologia , Fator 4 Nuclear de Hepatócito/fisiologia , Proteínas de Homeodomínio/fisiologia , Humanos , Regeneração Hepática/fisiologia , Fatores de Transcrição SOXF/fisiologia , Fatores de Transcrição/fisiologia
3.
Cancer Res ; 80(15): 3116-3129, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32518203

RESUMO

Kaposi sarcoma is a tumor caused by Kaposi sarcoma herpesvirus (KSHV) infection and is thought to originate from lymphatic endothelial cells (LEC). While KSHV establishes latency in virtually all susceptible cell types, LECs support spontaneous expression of oncogenic lytic genes, high viral genome copies, and release of infectious virus. It remains unknown the contribution of spontaneous virus production to the expansion of KSHV-infected tumor cells and the cellular factors that render the lymphatic environment unique to KSHV life cycle. We show here that expansion of the infected cell population, observed in LECs, but not in blood endothelial cells, is dependent on the spontaneous virus production from infected LECs. The drivers of lymphatic endothelium development, SOX18 and PROX1, regulated different steps of the KSHV life cycle. SOX18 enhanced the number of intracellular viral genome copies and bound to the viral origins of replication. Genetic depletion or chemical inhibition of SOX18 caused a decrease of KSHV genome copy numbers. PROX1 interacted with ORF50, the viral initiator of lytic replication, and bound to the KSHV genome in the promoter region of ORF50, increasing its transactivation activity and KSHV spontaneous lytic gene expression and infectious virus release. In Kaposi sarcoma tumors, SOX18 and PROX1 expression correlated with latent and lytic KSHV protein expression. These results demonstrate the importance of two key transcriptional drivers of LEC fate in the regulation of the tumorigenic KSHV life cycle. Moreover, they introduce molecular targeting of SOX18 as a potential novel therapeutic avenue in Kaposi sarcoma. SIGNIFICANCE: SOX18 and PROX1, central regulators of lymphatic development, are key factors for KSHV genome maintenance and lytic cycle in lymphatic endothelial cells, supporting Kaposi sarcoma tumorigenesis and representing attractive therapeutic targets.


Assuntos
Transformação Celular Viral/genética , Herpesvirus Humano 8/fisiologia , Proteínas de Homeodomínio/fisiologia , Fatores de Transcrição SOXF/fisiologia , Sarcoma de Kaposi/genética , Proteínas Supressoras de Tumor/fisiologia , Replicação Viral/genética , Carcinogênese/genética , Células Cultivadas , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Células Endoteliais/virologia , Regulação Viral da Expressão Gênica/genética , Genoma Viral/genética , Células HEK293 , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/patogenicidade , Proteínas de Homeodomínio/genética , Humanos , Sistema Linfático/metabolismo , Sistema Linfático/patologia , Sistema Linfático/virologia , Fatores de Transcrição SOXF/genética , Sarcoma de Kaposi/patologia , Sarcoma de Kaposi/virologia , Proteínas Supressoras de Tumor/genética
4.
Eur Rev Med Pharmacol Sci ; 23(21): 9304-9312, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31773697

RESUMO

OBJECTIVE: To investigate the regulatory effect of microRNA-616 (miRNA-616) on cellular behaviors of bladder cancer and the potential mechanism. PATIENTS AND METHODS: The expressions of miRNA-616 and SOX7 in bladder cancer tissues and cell lines were examined by quantitative Real Time-Polymerase Chain Reaction (qRT-PCR). The relationship between miRNA-616 and SOX7 was assessed through Dual-Luciferase Reporter Gene Assay. The regulatory effects of miRNA-616 and SOX7 on cellular behaviors of bladder cancer were evaluated through cell counting kit-8 (CCK-8), colony formation, transwell migration assay, and flow cytometry. RESULTS: MiRNA-616 was upregulated, whereas SOX7 was downregulated in bladder cancer tissues and cell lines. The silence of miRNA-616 attenuated the proliferative and migratory abilities, arrested cell cycle progression in the G2 phase, and stimulated apoptosis in UMUC3 and T24 cells. SOX7 was the target gene of miRNA-616, and its level was negatively regulated by miRNA-616. The knockdown of SOX7 enhanced the proliferative and migratory abilities, and attenuated apoptosis of bladder cancer cells. CONCLUSIONS: MiRNA-616 accelerates bladder cancer cells to proliferate and migrate and inhibits apoptosis by downregulating SOX7. MiRNA-616/SOX7 may be potential therapeutic targets for bladder cancer.


Assuntos
Apoptose/fisiologia , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , MicroRNAs/fisiologia , Fatores de Transcrição SOXF/fisiologia , Neoplasias da Bexiga Urinária/genética , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Progressão da Doença , Regulação para Baixo/genética , Técnicas de Silenciamento de Genes , Humanos , MicroRNAs/antagonistas & inibidores , MicroRNAs/biossíntese , Fatores de Transcrição SOXF/biossíntese , Fatores de Transcrição SOXF/genética , Regulação para Cima/genética
5.
Sci Rep ; 9(1): 15495, 2019 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-31664088

RESUMO

The yin and yang of female fertility is a complicated issue; large numbers of women/couples desire fertility and seek assisted reproduction intervention to achieve conception, while others seek to prevent pregnancy. Understanding specific molecules which control endometrial-embryo interactions is essential for both facilitating and preventing pregnancy. SOX17 has recently emerged as an important transcription factor involved in endometrial receptivity and embryo implantation. However, studies to date have examined mouse models of pregnancy which do not necessarily translate to the human. Demonstration of a role for 'implantation factors' in a human system is critical to provide a rationale for in depth clinical investigation and targeting of such factors. We demonstrate that SOX17is present within the receptive human endometrium and is up-regulated within human endometrial epithelial cells by combined estrogen & progesterone, the hormonal milieu during the receptive window. SOX17 localizes to the point of adhesive contact between human endometrial epithelial cells and a human 'embryo mimic' model (trophectodermal spheroid). Targeting SOX17 in endometrial epithelial cells using CRISPR/Cas9 knockdown or a SOX-F family inhibitor, MCC177, significantly inhibited adhesion of an trophectodermal spheroids to the epithelial cells thereby preventing 'implantation'. These data confirm the important role of endometrial SOX17 in human endometrial receptivity and embryo implantation.


Assuntos
Implantação do Embrião/fisiologia , Endométrio/fisiologia , Fatores de Transcrição SOXF/fisiologia , Estrogênios/fisiologia , Feminino , Técnicas de Silenciamento de Genes , Humanos , Gravidez , Progesterona/fisiologia , Fatores de Transcrição SOXF/genética
6.
Oncogene ; 38(34): 6196-6210, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31332289

RESUMO

Apoptosis of cancer cells occurs by a complex gene regulatory network. Here we showed that SOX7 was significantly downregulated in different cancer types, especially in lung and breast cancers. Low expression of SOX7 was associated with advantage stage of cancer with shorter overall survival. Cancer cells with loss of SOX7 promoted cell survival and colony formation, suppressed cellular apoptosis and produced a drug resistant phenotype against a variety of chemo/targeting therapeutic agents. Mechanistically, SOX7 induced cellular apoptosis through upregulation of genes associated with both P38 and apoptotic signaling pathway, as well as preventing the proteasome mediated degradation of pro-apoptotic protein BIM. Treatment of either a proteasome inhibitor MG132 or bortezomib, or with a p-ERK/MEK inhibitor U0126 attenuate the SOX7 promoted BIM degradation. We identified Panobinostat, an FDA approved pan-HDAC inhibitor, could elevate and restore SOX7 expression in SOX7 silenced lung cancer cells. Taken together, these data revealed an unappreciated role of SOX7 in regulation of cellular apoptosis through control of MAPK/ERK-BIM signaling.


Assuntos
Apoptose/genética , Sistema de Sinalização das MAP Quinases/fisiologia , Neoplasias/patologia , Fatores de Transcrição SOXF/fisiologia , Animais , Proteína 11 Semelhante a Bcl-2/genética , Proteína 11 Semelhante a Bcl-2/metabolismo , Sobrevivência Celular/genética , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Sistema de Sinalização das MAP Quinases/genética , Masculino , Camundongos , Camundongos SCID , Neoplasias/genética , Neoplasias/metabolismo , Fatores de Transcrição SOXF/genética , Células Tumorais Cultivadas
7.
Neuromolecular Med ; 21(4): 325-343, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31055715

RESUMO

Intracranial aneurysms (IA) are local dilatations in cerebral arteries that predominantly affect the circle of Willis. Occurring in approximately 2-5% of adults, these weakened areas are susceptible to rupture, leading to subarachnoid hemorrhage (SAH), a type of hemorrhagic stroke. Due to its early age of onset and poor prognosis, SAH accounts for > 25% of years lost for all stroke victims under the age of 65. In this review, we describe the cerebrovascular pathology associated with intracranial aneurysms. To understand IA genetics, we summarize syndromes with elevated incidence, genome-wide association studies (GWAS), whole exome studies on IA-affected families, and recent research that established definitive roles for Thsd1 (Thrombospondin Type 1 Domain Containing Protein 1) and Sox17 (SRY-box 17) in IA using genetically engineered mouse models. Lastly, we discuss the underlying molecular mechanisms of IA, including defects in vascular endothelial and smooth muscle cells caused by dysfunction in mechanotransduction, Thsd1/FAK (Focal Adhesion Kinase) signaling, and the Transforming Growth Factor ß (TGF-ß) pathway. As illustrated by THSD1 research, cell adhesion may play a significant role in IA.


Assuntos
Aneurisma Intracraniano , Aneurisma Roto/complicações , Animais , Arterite/complicações , Arterite/patologia , Estudos de Casos e Controles , Artérias Cerebrais/ultraestrutura , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Adesões Focais , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Hemorreologia , Humanos , Incidência , Aneurisma Intracraniano/complicações , Aneurisma Intracraniano/epidemiologia , Aneurisma Intracraniano/genética , Aneurisma Intracraniano/patologia , Mamíferos , Mecanotransdução Celular , Camundongos , Miócitos de Músculo Liso/patologia , Fatores de Transcrição SOXF/fisiologia , Hemorragia Subaracnóidea/etiologia , Síndrome , Trombospondinas/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Sequenciamento do Exoma , Peixe-Zebra
8.
Prostate ; 79(4): 370-378, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30488457

RESUMO

BACKGROUND: PSMA expression in the prostate epithelium is controlled by a cis-element, PSMA enhancer (PSME). PSME contains multiple binding sites for Sox proteins, and in this study, we identified Sox7 protein as a negative regulator of PSMA expression through its interaction with PSME. METHODS: The statistical correlation between Sox7 and PSMA mRNA expression was evaluated using five prostate cancer studies from cBioportal. In vitro and in vivo interaction between Sox7 and PSME was evaluated by chromatin immunoprecipitation (ChIP), electrophoretic mobility shift assay (EMSA), and luciferase reporter assay. Synthetic oligonucleotides were generated to define the sites in PSME that interact with Sox7 protein. Sox7 mutants were generated to identify the region of this protein required to regulate PSMA expression. Sox7 was also stably expressed in LNCaP/C4-2 and 22Rv1 cells to validate the regulation of PSMA expression by Sox7 in vivo. RESULTS: Sox7 mRNA expression negatively correlated with PSMA/FOLH1 and PSMAL/FOLH1B mRNA expression in Broad/Cornell, TCGA and MSKCC studies, but not in two studies containing only metastatic prostate tumors. PC-3 cells mostly expressed the 48.5 KDa isoform 2 of Sox7, and the depletion of this isoform did not restore PSMA expression. Ectopic expression of canonical, wild-type Sox7 in C4-2 and 22Rv1 cells suppressed PSMA protein expression. ChIP assay revealed that canonical Sox7 protein preferentially interacts with PSME in vivo, and EMSA identified the SOX box sites #2 and #4 in PSME as required for its interaction. Sox7 was capable of directly binding to PSME and suppressed PSME-mediated transcription. The NLS regions of Sox7, but not its ß-catenin interacting motif, are essential for this suppressing activity. Furthermore, restoration of wild-type Sox7 expression but not Sox7-NLS mutant in Sox7-null prostate cancer cell lines suppressed PSMA expression. CONCLUSIONS: The inactivation of canonical Sox7 is responsible for the upregulated expression of PSMA in non-metastatic prostate cancer.


Assuntos
Antígenos de Superfície/genética , Elementos Facilitadores Genéticos/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Glutamato Carboxipeptidase II/genética , Próstata/metabolismo , Neoplasias da Próstata/metabolismo , Fatores de Transcrição SOXF/fisiologia , Sequência de Aminoácidos , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Humanos , Masculino , Neoplasias da Próstata/química , RNA Mensageiro/análise , Fatores de Transcrição SOXF/química , Via de Sinalização Wnt/fisiologia
9.
Ann Anat ; 219: 25-34, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29842991

RESUMO

While both blood and lymphatic vessels transport fluids and thus share many similarities, they also show functional and structural differences, which can be used to differentiate them. Specific visualization of lymphatic vessels has historically been and still is a pivot point in lymphatic research. Many of the proteins that are investigated by molecular biologists in lymphatic research have been defined as marker molecules, i.e. to visualize and distinguish lymphatic endothelial cells (LECs) from other cell types, most notably from blood vascular endothelial cells (BECs) and cells of the hematopoietic lineage. Among the factors that drive the developmental differentiation of lymphatic structures from venous endothelium, Prospero homeobox protein 1 (PROX1) is the master transcriptional regulator. PROX1 maintains lymphatic identity also in the adult organism and thus is a universal LEC marker. Vascular endothelial growth factor receptor-3 (VEGFR-3) is the major tyrosine kinase receptor that drives LEC proliferation and migration. The major activator for VEGFR-3 is vascular endothelial growth factor-C (VEGF-C). However, before VEGF-C can signal, it needs to be proteolytically activated by an extracellular protein complex comprised of Collagen and calcium binding EGF domains 1 (CCBE1) protein and the protease A disintegrin and metallopeptidase with thrombospondin type 1 motif 3 (ADAMTS3). This minireview attempts to give an overview of these and a few other central proteins that scientific inquiry has linked specifically to the lymphatic vasculature. It is limited in scope to a brief description of their main functions, properties and developmental roles.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Sistema Linfático/fisiologia , Receptores de Superfície Celular/fisiologia , Fatores de Transcrição/fisiologia , Animais , Fator II de Transcrição COUP/fisiologia , Fatores de Transcrição Forkhead/fisiologia , Proteínas de Homeodomínio/fisiologia , Humanos , Linfangiogênese/fisiologia , Sistema Linfático/crescimento & desenvolvimento , Fatores de Transcrição SOXF/fisiologia , Transdução de Sinais , Proteínas Supressoras de Tumor/fisiologia , Fator C de Crescimento do Endotélio Vascular/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/fisiologia
10.
Biol Reprod ; 99(3): 578-589, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29635272

RESUMO

In mouse conceptus, two yolk-sac membranes, the parietal endoderm (PE) and visceral endoderm (VE), are involved in protecting and nourishing early-somite-stage embryos prior to the establishment of placental circulation. Both PE and VE membranes are tightly anchored to the marginal edge of the developing placental disk, in which the extraembryonic endoderm (marginal zone endoderm: ME) shows the typical flat epithelial morphology intermediate between those of PE and VE in vivo. However, the molecular characteristics and functions of the ME in mouse placentation remain unclear. Here, we show that SOX17, not SOX7, is continuously expressed in the ME cells, whereas both SOX17 and SOX7 are coexpressed in PE cells, by at least 10.5 days postconception. The Sox17-null conceptus, but not the Sox7-null one, showed the ectopic appearance of squamous VE-like epithelial cells in the presumptive ME region, together with reduced cell density and aberrant morphology of PE cells. Such aberrant ME formation in the Sox17-null extraembryonic endoderm was not rescued by the chimeric embryo replaced with the wild-type gut endoderm by the injection of wild-type ES cells into the Sox17-null blastocyst, suggesting the cell autonomous defects in the extraembryonic endoderm of Sox17-null concepti. These findings provide direct evidence of the crucial roles of SOX17 in proper formation and maintenance of the ME region, highlighting a novel entry point to understand the in vivo VE-to-PE transition in the marginal edge of developing placenta.


Assuntos
Desenvolvimento Embrionário/fisiologia , Endoderma/fisiologia , Proteínas HMGB/fisiologia , Placentação/fisiologia , Fatores de Transcrição SOXF/fisiologia , Saco Vitelino/fisiologia , Animais , Proliferação de Células , Feminino , Expressão Gênica , Genótipo , Proteínas HMGB/deficiência , Proteínas HMGB/genética , Masculino , Camundongos , Camundongos Knockout , Gravidez , Fatores de Transcrição SOXF/deficiência , Fatores de Transcrição SOXF/genética
11.
DNA Cell Biol ; 37(2): 126-132, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29271667

RESUMO

The Wnt signaling is involved in angiogenesis and tumor development. ß-catenin is the core component of the Wnt pathway, which mediates oncogenic transcription and regulated by a series of proteins. Sex-determining region Y-box 7 (SOX7) is a member of high-mobility-group transcription factor family, which inhibits oncogenic Wnt signaling in lots of tumor cells with unknown mechanism. By coimmunoprecipitation (co-IP) and super Topflash reporter assay, SOX7 can bind ß-catenin and inhibit ß-catenin/T cell factor (TCF)-mediated transcription. Meanwhile, B cell lymphoma 9 (BCL9) drives Wnt signaling path through direct binding-mediated ß-catenin. Finally, we found that SOX7 inhibits oncogenic ß-catenin-mediated transcription by disrupting the ß-catenin/BCL9 interaction. Mechanistically, SOX7 compete with BCL9 to bind ß-catenin. Our results show SOX7 inhibited Wnt signaling as suppressor and could be an important target for anticancer therapy.


Assuntos
Proteínas de Neoplasias/metabolismo , Fatores de Transcrição SOXF/fisiologia , beta Catenina/metabolismo , Proliferação de Células , Células HEK293 , Humanos , Ligação Proteica , Fatores de Transcrição , Transcrição Gênica , Via de Sinalização Wnt
12.
Development ; 144(10): 1887-1895, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28512199

RESUMO

SOX family proteins SOX2 and SOX18 have been reported as being essential in determining hair follicle type; however, the role they play during development remains unclear. Here, we demonstrate that Sox18 regulates the normal differentiation of the dermal papilla of all hair types. In guard (primary) hair dermal condensate (DC) cells, we identified transient Sox18 in addition to SOX2 expression at E14.5, which allowed fate tracing of primary DC cells until birth. Similarly, expression of Sox18 was detected in the DC cells of secondary hairs at E16.5 and in tertiary hair at E18.5. Dominant-negative Sox18 mutation (opposum) did not prevent DC formation in any hair type. However, it affected dermal papilla differentiation, restricting hair formation especially in secondary and tertiary hairs. This Sox18 mutation also prevented neonatal dermal cells or dermal papilla spheres from inducing hair in regeneration assays. Microarray expression studies identified WNT5A and TNC as potential downstream effectors of SOX18 that are important for epidermal WNT signalling. In conclusion, SOX18 acts as a mesenchymal molecular switch necessary for the formation and function of the dermal papilla in all hair types.


Assuntos
Diferenciação Celular/genética , Folículo Piloso/embriologia , Cabelo/embriologia , Fatores de Transcrição SOXF/fisiologia , Animais , Derme/embriologia , Derme/metabolismo , Embrião de Mamíferos , Células Epidérmicas , Epiderme/embriologia , Feminino , Genes Dominantes , Genes de Troca/fisiologia , Cabelo/metabolismo , Folículo Piloso/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Fatores de Transcrição SOXF/genética
13.
Circulation ; 135(25): 2505-2523, 2017 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-28381471

RESUMO

BACKGROUND: The mechanisms underlying the dedifferentiation and lineage conversion of adult human fibroblasts into functional endothelial cells have not yet been fully defined. Furthermore, it is not known whether fibroblast dedifferentiation recapitulates the generation of multipotent progenitors during embryonic development, which give rise to endothelial and hematopoietic cell lineages. Here we established the role of the developmental transcription factor SOX17 in regulating the bilineage conversion of fibroblasts by the generation of intermediate progenitors. METHODS: CD34+ progenitors were generated after the dedifferentiation of human adult dermal fibroblasts by overexpression of pluripotency transcription factors. Sorted CD34+ cells were transdifferentiated into induced endothelial cells and induced erythroblasts using lineage-specific growth factors. The therapeutic potential of the generated cells was assessed in an experimental model of myocardial infarction. RESULTS: Induced endothelial cells expressed specific endothelial cell surface markers and also exhibited the capacity for cell proliferation and neovascularization. Induced erythroblasts expressed erythroid surface markers and formed erythroid colonies. Endothelial lineage conversion was dependent on the upregulation of the developmental transcription factor SOX17, whereas suppression of SOX17 instead directed the cells toward an erythroid fate. Implantation of these human bipotential CD34+ progenitors into nonobese diabetic/severe combined immunodeficiency (NOD-SCID) mice resulted in the formation of microvessels derived from human fibroblasts perfused with mouse and human erythrocytes. Endothelial cells generated from human fibroblasts also showed upregulation of telomerase. Cell implantation markedly improved vascularity and cardiac function after myocardial infarction without any evidence of teratoma formation. CONCLUSIONS: Dedifferentiation of fibroblasts to intermediate CD34+ progenitors gives rise to endothelial cells and erythroblasts in a SOX17-dependent manner. These findings identify the intermediate CD34+ progenitor state as a critical bifurcation point, which can be tuned to generate functional blood vessels or erythrocytes and salvage ischemic tissue.


Assuntos
Antígenos CD34/fisiologia , Desdiferenciação Celular/fisiologia , Células Endoteliais/fisiologia , Eritroblastos/fisiologia , Fibroblastos/fisiologia , Fatores de Transcrição SOXF/fisiologia , Células-Tronco/fisiologia , Animais , Células Cultivadas , Humanos , Recém-Nascido , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
14.
J Hepatol ; 67(1): 72-83, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28237397

RESUMO

BACKGROUND & AIMS: Cholangiocarcinoma (CCA) is a biliary malignancy linked to genetic and epigenetic abnormalities, such as hypermethylation of SOX17 promoter. Here, the role of SOX17 in cholangiocyte differentiation and cholangiocarcinogenesis was studied. METHODS: SOX17 expression/function was evaluated along the differentiation of human induced pluripotent stem cells (iPSC) into cholangiocytes, in the dedifferentiation process of normal human cholangiocytes (NHC) in culture and in cholangiocarcinogenesis. Lentiviruses for SOX17 overexpression or knockdown were used. Gene expression and DNA methylation profiling were performed. RESULTS: SOX17 expression is induced in the last stage of cholangiocyte differentiation from iPSC and regulates the acquisition of biliary markers. SOX17 becomes downregulated in NHC undergoing dedifferentiation; experimental SOX17 knockdown in differentiated NHC downregulated biliary markers and promoted baseline and Wnt-dependent proliferation. SOX17 expression is lower in human CCA than in healthy tissue, which correlates with worse survival after tumor resection. In CCA cells, SOX17 overexpression decreased their tumorigenic capacity in murine xenograft models, which was related to increased oxidative stress and apoptosis. In contrast, SOX17 overexpression in NHC did not affect their survival but inhibited their baseline proliferation. In CCA cells, SOX17 inhibited migration, anchorage-independent growth and Wnt/ß-catenin-dependent proliferation, and restored the expression of biliary markers and primary cilium length. In human CCA, SOX17 promoter was found hypermethylated and its expression inversely correlates with the methylation grade. In NHC, Wnt3a decreased SOX17 expression in a DNMT-dependent manner, whereas in CCA, DNMT1 inhibition or silencing upregulated SOX17. CONCLUSIONS: SOX17 regulates the differentiation and maintenance of the biliary phenotype and functions as a tumor suppressor for CCA, being a potential prognostic marker and a promising therapeutic target. LAY SUMMARY: Understanding the molecular mechanisms involved in the pathogenesis of CCA is key in finding new valuable diagnostic and prognostic biomarkers, as well as therapeutic targets. This study provides evidence that SOX17 regulates the differentiation and maintenance of the biliary phenotype, and its downregulation promotes their tumorigenic transformation. SOX17 acts as a tumor suppressor in CCA and its genetic, molecular and/or pharmacological restoration may represent a new promising therapeutic strategy. Moreover, SOX17 expression correlates with the outcome of patients after tumor resection, being a potential prognostic biomarker.


Assuntos
Neoplasias dos Ductos Biliares/etiologia , Ductos Biliares/patologia , Colangiocarcinoma/etiologia , Fatores de Transcrição SOXF/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Diferenciação Celular , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Fatores de Transcrição SOXF/análise , Fatores de Transcrição SOXF/genética
15.
Circ Res ; 119(7): 839-52, 2016 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-27528602

RESUMO

RATIONALE: Vascular endothelial growth factor (VEGF) signaling is a key pathway for angiogenesis and requires highly coordinated regulation. Although the Notch pathway-mediated suppression of excessive VEGF activity via negative feedback is well known, the positive feedback control for augmenting VEGF signaling remains poorly understood. Transcription factor Sox17 is indispensable for angiogenesis, but its association with VEGF signaling is largely unknown. The contribution of other Sox members to angiogenesis also remains to be determined. OBJECTIVE: To reveal the genetic interaction of Sox7, another Sox member, with Sox17 in developmental angiogenesis and their functional relationship with VEGF signaling. METHODS AND RESULTS: Sox7 is expressed specifically in endothelial cells and its global and endothelial-specific deletion resulted in embryonic lethality with severely impaired angiogenesis in mice, substantially overlapping with Sox17 in both expression and function. Interestingly, compound heterozygosity for Sox7 and Sox17 phenocopied vascular defects of Sox7 or Sox17 homozygous knockout, indicating that the genetic cooperation of Sox7 and Sox17 is sensitive to their combined gene dosage. VEGF signaling upregulated both Sox7 and Sox17 expression in angiogenesis via mTOR pathway. Furthermore, Sox7 and Sox17 promoted VEGFR2 (VEGF receptor 2) expression in angiogenic vessels, suggesting a positive feedback loop between VEGF signaling and SoxF. CONCLUSIONS: Our findings demonstrate that SoxF transcription factors are indispensable players in developmental angiogenesis by acting as positive feedback regulators of VEGF signaling.


Assuntos
Células Endoteliais da Veia Umbilical Humana/metabolismo , Neovascularização Fisiológica/fisiologia , Fatores de Transcrição SOXF/fisiologia , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Técnicas de Cultura , Feminino , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Gravidez
16.
Dev Biol ; 414(2): 219-27, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27102016

RESUMO

The importance of canonical Wnt signaling to murine uterine development is well established. Mouse models in which uterine-specific Wnt ligands, ß-catenin, or Lef1 are disrupted result in failure of postnatal endometrial gland development. Sox17 is a transcription factor characterized in numerous tissues as an antagonist of Wnt signaling. Thus, we hypothesized that conditional ablation of Sox17 would lead to hyperproliferation of endometrial glands in mice. Contrary to our prediction, disruption of Sox17 in epithelial and stromal compartments led to inhibition of endometrial adenogenesis and a loss of reproductive capacity. Epithelium-specific Sox17 disruption resulted in normal adenogenesis although reproductive capacity remained impaired. These findings suggest that non-epithelial, Sox17-positive cells are necessary for adenogenesis and that glands require Sox17 to properly function. To our knowledge, these findings are the first to implicate Sox17 in endometrial gland formation and reproductive success. The data presented herein underscore the importance of studying Sox17 in uterine homeostasis and function.


Assuntos
Endométrio/crescimento & desenvolvimento , Células Epiteliais/fisiologia , Glândulas Exócrinas/crescimento & desenvolvimento , Proteínas HMGB/fisiologia , Fatores de Transcrição SOXF/fisiologia , Animais , Endométrio/metabolismo , Endométrio/patologia , Glândulas Exócrinas/metabolismo , Feminino , Proteínas HMGB/deficiência , Proteínas HMGB/genética , Fator 3-beta Nuclear de Hepatócito/biossíntese , Fator 3-beta Nuclear de Hepatócito/genética , Homeostase , Hiperplasia , Infertilidade Feminina/genética , Infertilidade Feminina/patologia , Proteínas Luminescentes/análise , Fator 1 de Ligação ao Facilitador Linfoide/biossíntese , Fator 1 de Ligação ao Facilitador Linfoide/genética , Camundongos , Camundongos Knockout , Gravidez , Fatores de Transcrição SOXF/deficiência , Fatores de Transcrição SOXF/genética , Organismos Livres de Patógenos Específicos , Células Estromais/fisiologia , Proteínas Wnt/biossíntese , Proteínas Wnt/genética , Via de Sinalização Wnt/fisiologia
17.
Cancer Lett ; 375(1): 100-107, 2016 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-26944317

RESUMO

SOX7 has been recently recognized as a tumor suppressor belonging to the SOX (SRY-related HMG-box) family of a transcription factor. However, its role in human gliomas is unknown. Our study showed that SOX7 expression was significantly downregulated in human gliomas. Statistical analysis showed that SOX7 suppression was associated with higher histological grades of tumors in glioma tissues. SOX7 could suppress tumor properties both in vivo and in vitro, and depletion of the HMG domain abolishes its tumor suppressive roles. In vitro assays demonstrated that SOX7 could downregulate Wnt/ß-catenin transcription and decrease the expression of Cyclin D1 and c-Myc, while the mutant SOX7 lost these functions. These results suggested that the HMG-box is a key domain of SOX7 for negatively regulating the Wnt/ß-catenin signaling pathway when functioning as a tumor suppressor in a glioma.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Fatores de Transcrição SOXF/fisiologia , Via de Sinalização Wnt , Adulto , Animais , Sítios de Ligação , Carcinogênese , Linhagem Celular Tumoral , Proliferação de Células , Regulação para Baixo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos Nus , Pessoa de Meia-Idade , Transplante de Neoplasias , Estrutura Terciária de Proteína
18.
Tumour Biol ; 37(9): 11753-11762, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27022736

RESUMO

Interleukin-22 (IL-22) is an inflammatory cytokine mainly produced by activated Th17 and Th22 cells. The data presented here demonstrate that IL-22 induced the migration and invasion of papillary thyroid cancer (PTC) cells. MicroRNA expression analysis and functional studies indicated that IL-22-mediated migration and invasion is positively regulated by miR-595. Further mechanistic studies revealed that sex-determining region Y-box 17 (Sox17) is directly targeted by miR-595. We then demonstrated that IL-22 regulated migration and invasion of PTC cells via inhibiting Sox17 expression. Interestingly, in PTC cell lines and PTC tissues, expression of IL-22 and miR-595 was upregulated and Sox17 downregulated compared with normal thyroid, and their expression levels were closely correlated. Taken together, this present study suggests that IL-22 stimulation enhances the migration and invasion of PTC cells by regulating miR-595 and its target Sox17.


Assuntos
Carcinoma/patologia , Interleucinas/farmacologia , MicroRNAs/fisiologia , Fatores de Transcrição SOXF/fisiologia , Neoplasias da Glândula Tireoide/patologia , Carcinoma Papilar , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Humanos , Invasividade Neoplásica , Fatores de Transcrição SOXF/genética , Câncer Papilífero da Tireoide , Interleucina 22
19.
Nat Commun ; 6: 7739, 2015 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-26204127

RESUMO

Changes in cell fate and identity are essential for endothelial-to-haematopoietic transition (EHT), an embryonic process that generates the first adult populations of haematopoietic stem cells (HSCs) from hemogenic endothelial cells. Dissecting EHT regulation is a critical step towards the production of in vitro derived HSCs. Yet, we do not know how distinct endothelial and haematopoietic fates are parsed during the transition. Here we show that genes required for arterial identity function later to repress haematopoietic fate. Tissue-specific, temporally controlled, genetic loss of arterial genes (Sox17 and Notch1) during EHT results in increased production of haematopoietic cells due to loss of Sox17-mediated repression of haematopoietic transcription factors (Runx1 and Gata2). However, the increase in EHT can be abrogated by increased Notch signalling. These findings demonstrate that the endothelial haematopoietic fate switch is actively repressed in a population of endothelial cells, and that derepression of these programs augments haematopoietic output.


Assuntos
Vasos Sanguíneos/embriologia , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Fator de Transcrição GATA2/metabolismo , Proteínas HMGB/fisiologia , Hemangioblastos/fisiologia , Fatores de Transcrição SOXF/fisiologia , Animais , Feminino , Genes Reporter , Hematopoese , Camundongos , Gravidez , Receptor Notch1/metabolismo
20.
Oncotarget ; 6(11): 9099-112, 2015 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-25868860

RESUMO

Cancer stem cells (CSCs) and EMT-type cells, which share molecular characteristics with CSCs, have been believed to play critical roles in tumor metastasis. Although much progress has been garnered in elucidating the molecular pathways that trigger EMT, stemness and metastasis, a number of key mechanistic gaps remain elusive. In the study, miR-371-5p was obviously down-regulated in primary CRC tissues compared with matched adjacent normal mucosa and correlated significantly with differentiation, tumor size, lymphatic and liver metastases. MiR-371-5p could attenuate proliferation, invasion in vitro and metastasis in vivo in CRC cells. It also suppressed EMT by regulating Wnt/ß-catenin signaling and strongly decreased the CRC stemness phenotypes. Moreover, demethylation of SOX17 induced miR-371-5p expression and consequently suppressed its direct target SOX2 in CRC cells. MiR-371-5p was necessary for SOX17 mediated cancer-related traits and SOX2 was a functional target of miR-371-5p. A positive relationship between SOX17 and miR-371-5p expression and a negative one between miR-371-5p and SOX2 expression were observed in CRC cell lines and tissues. In conclusion, we identified miR-371-5p as an important "oncosuppressor" in CRC progression and elucidated a novel mechanism of the SOX17/miR-371-5p/SOX2 axis in the regulation of EMT, stemness and metastasis, which may be a potential therapeutic target.


Assuntos
Adenocarcinoma/fisiopatologia , Neoplasias Colorretais/fisiopatologia , Transição Epitelial-Mesenquimal/fisiologia , Metástase Neoplásica/fisiopatologia , Proteínas de Neoplasias/fisiologia , Células-Tronco Neoplásicas/citologia , Adenocarcinoma/patologia , Adenocarcinoma/secundário , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Mucosa Intestinal/metabolismo , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/secundário , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/fisiologia , Invasividade Neoplásica , Células-Tronco Pluripotentes/citologia , RNA Neoplásico , Fatores de Transcrição SOXB1/fisiologia , Fatores de Transcrição SOXF/fisiologia , Transdução de Sinais/fisiologia , Organismos Livres de Patógenos Específicos , Transdução Genética , Via de Sinalização Wnt
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA