Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Oncoimmunology ; 11(1): 2033433, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35111387

RESUMO

In classical Hodgkin lymphoma (cHL), the highly abundant CD4+ T cells in the vicinity of tumor cells are considered essential for tumor cell survival, but are ill-defined. Although they are activated, they consistently lack expression of activation marker CD26. In this study, we compared sorted CD4+CD26- and CD4+CD26+ T cells from cHL lymph node cell suspensions by RNA sequencing and T cell receptor variable gene segment usage analysis. This revealed that although CD4+CD26- T cells are antigen experienced, they have not clonally expanded. This may well be explained by the expression of exhaustion associated transcription factors TOX and TOX2, immune checkpoints PDCD1 and CD200, and chemokine CXCL13, which were amongst the 100 significantly enriched genes in comparison with the CD4+CD26+ T cells. Findings were validated in single-cell RNA sequencing data from an independent cohort. Interestingly, immunohistochemistry revealed predominant and high frequency of staining for TOX and TOX2 in the T cells attached to the tumor cells. In conclusion, the dominant CD4+CD26- T cell population in cHL is antigen experienced, polyclonal, and exhausted. This population is likely a main contributor to the very high response rates to immune checkpoint inhibitors in cHL.


Assuntos
Linfócitos T CD4-Positivos , Proteínas HMGB , Proteínas de Grupo de Alta Mobilidade , Doença de Hodgkin , Dipeptidil Peptidase 4/imunologia , Proteínas HMGB/biossíntese , Proteínas HMGB/imunologia , Proteínas de Grupo de Alta Mobilidade/biossíntese , Proteínas de Grupo de Alta Mobilidade/imunologia , Doença de Hodgkin/genética , Doença de Hodgkin/imunologia , Doença de Hodgkin/metabolismo , Humanos , Linfonodos/patologia , Fatores de Transcrição/genética
2.
Sci Rep ; 9(1): 11953, 2019 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-31420575

RESUMO

The endocardium is the endothelial component of the vertebrate heart and plays a key role in heart development. Where, when, and how the endocardium segregates during embryogenesis have remained largely unknown, however. We now show that Nkx2-5+ cardiac progenitor cells (CPCs) that express the Sry-type HMG box gene Sox17 from embryonic day (E) 7.5 to E8.5 specifically differentiate into the endocardium in mouse embryos. Although Sox17 is not essential or sufficient for endocardium fate, it can bias the fate of CPCs toward the endocardium. On the other hand, Sox17 expression in the endocardium is required for heart development. Deletion of Sox17 specifically in the mesoderm markedly impaired endocardium development with regard to cell proliferation and behavior. The proliferation of cardiomyocytes, ventricular trabeculation, and myocardium thickening were also impaired in a non-cell-autonomous manner in the Sox17 mutant, likely as a consequence of down-regulation of NOTCH signaling. An unknown signal, regulated by Sox17 and required for nurturing of the myocardium, is responsible for the reduction in NOTCH-related genes in the mutant embryos. Our results thus provide insight into differentiation of the endocardium and its role in heart development.


Assuntos
Diferenciação Celular , Embrião de Mamíferos/embriologia , Endocárdio/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas HMGB/biossíntese , Fatores de Transcrição SOXF/biossíntese , Transdução de Sinais , Células-Tronco/metabolismo , Animais , Embrião de Mamíferos/citologia , Endocárdio/citologia , Proteínas HMGB/genética , Mesoderma/citologia , Mesoderma/embriologia , Camundongos , Camundongos Transgênicos , Receptores Notch/genética , Receptores Notch/metabolismo , Fatores de Transcrição SOXF/genética , Células-Tronco/citologia
3.
Proc Natl Acad Sci U S A ; 113(38): 10583-8, 2016 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-27601662

RESUMO

The transcriptional repressor Capicua (Cic) controls tissue patterning and restricts organ growth, and has been recently implicated in several cancers. Cic has emerged as a primary sensor of signaling downstream of the receptor tyrosine kinase (RTK)/extracellular signal-regulated kinase (ERK) pathway, but how Cic activity is regulated in different cellular contexts remains poorly understood. We found that the kinase Minibrain (Mnb, ortholog of mammalian DYRK1A), acting through the adaptor protein Wings apart (Wap), physically interacts with and phosphorylates the Cic protein. Mnb and Wap inhibit Cic function by limiting its transcriptional repressor activity. Down-regulation of Cic by Mnb/Wap is necessary for promoting the growth of multiple organs, including the wings, eyes, and the brain, and for proper tissue patterning in the wing. We have thus uncovered a previously unknown mechanism of down-regulation of Cic activity by Mnb and Wap, which operates independently from the ERK-mediated control of Cic. Therefore, Cic functions as an integrator of upstream signals that are essential for tissue patterning and organ growth. Finally, because DYRK1A and CIC exhibit, respectively, prooncogenic vs. tumor suppressor activities in human oligodendroglioma, our results raise the possibility that DYRK1A may also down-regulate CIC in human cells.


Assuntos
Padronização Corporal/genética , Proteínas de Drosophila/genética , Drosophila/genética , Proteínas HMGB/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/genética , Proteínas Repressoras/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Drosophila/crescimento & desenvolvimento , Proteínas de Drosophila/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Proteínas HMGB/biossíntese , Humanos , Neoplasias/genética , Fosforilação , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Repressoras/biossíntese , Asas de Animais/crescimento & desenvolvimento , Quinases Dyrk
4.
PLoS Genet ; 11(12): e1005634, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26683696

RESUMO

Epithelial renewal in the Drosophila intestine is orchestrated by Intestinal Stem Cells (ISCs). Following damage or stress the intestinal epithelium produces ligands that activate the epidermal growth factor receptor (EGFR) in ISCs. This promotes their growth and division and, thereby, epithelial regeneration. Here we demonstrate that the HMG-box transcriptional repressor, Capicua (Cic), mediates these functions of EGFR signaling. Depleting Cic in ISCs activated them for division, whereas overexpressed Cic inhibited ISC proliferation and midgut regeneration. Epistasis tests showed that Cic acted as an essential downstream effector of EGFR/Ras signaling, and immunofluorescence showed that Cic's nuclear localization was regulated by EGFR signaling. ISC-specific mRNA expression profiling and DNA binding mapping using DamID indicated that Cic represses cell proliferation via direct targets including string (Cdc25), Cyclin E, and the ETS domain transcription factors Ets21C and Pointed (pnt). pnt was required for ISC over-proliferation following Cic depletion, and ectopic pnt restored ISC proliferation even in the presence of overexpressed dominant-active Cic. These studies identify Cic, Pnt, and Ets21C as critical downstream effectors of EGFR signaling in Drosophila ISCs.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Receptores ErbB/genética , Proteínas HMGB/genética , Proteínas do Tecido Nervoso/genética , Proteínas Proto-Oncogênicas c-ets/genética , Proteínas Proto-Oncogênicas/genética , Receptores de Peptídeos de Invertebrados/genética , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Animais , Proliferação de Células/genética , Proteínas de Ligação a DNA/biossíntese , Drosophila/genética , Proteínas de Drosophila/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Proteínas HMGB/biossíntese , Intestinos/citologia , Intestinos/crescimento & desenvolvimento , Proteínas do Tecido Nervoso/biossíntese , Proteínas Proto-Oncogênicas/biossíntese , RNA Mensageiro/biossíntese , Proteínas Repressoras/biossíntese , Transdução de Sinais/genética , Células-Tronco/citologia , Fatores de Transcrição/biossíntese
5.
Nat Cell Biol ; 16(12): 1146-56, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25419850

RESUMO

Gastrulation leads to three germ layers--ectoderm, mesoderm and endoderm--that are separated by two basement membranes. In the mouse embryo, the emergent gut endoderm results from the widespread intercalation of cells of two distinct origins: pluripotent epiblast-derived definitive endoderm (DE) and extra-embryonic visceral endoderm (VE). Here we image the trajectory of prospective DE cells before intercalating into the VE epithelium. We show that the transcription factor SOX17, which is activated in prospective DE cells before intercalation, is necessary for gut endoderm morphogenesis and the assembly of the basement membrane that separates gut endoderm from mesoderm. Our results mechanistically link gut endoderm morphogenesis and germ layer segregation, two central and conserved features of gastrulation.


Assuntos
Endoderma/embriologia , Camadas Germinativas/embriologia , Proteínas HMGB/metabolismo , Mesoderma/embriologia , Morfogênese/fisiologia , Fatores de Transcrição SOXF/metabolismo , Animais , Membrana Basal/citologia , Membrana Basal/embriologia , Caderinas/biossíntese , Diferenciação Celular , Embrião de Mamíferos , Epitélio/embriologia , Matriz Extracelular/metabolismo , Proteínas da Matriz Extracelular/biossíntese , Fibronectinas/metabolismo , Gastrulação , Proteínas de Fluorescência Verde/genética , Proteínas HMGB/biossíntese , Fator 3-beta Nuclear de Hepatócito/genética , Camundongos , Camundongos Transgênicos , Morfogênese/genética , Imagem Óptica/métodos , Fatores de Transcrição SOXF/biossíntese
6.
Dev Cell ; 29(4): 454-67, 2014 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-24835466

RESUMO

Cells of the inner cell mass (ICM) of the mouse blastocyst differentiate into the pluripotent epiblast or the primitive endoderm (PrE), marked by the transcription factors NANOG and GATA6, respectively. To investigate the mechanistic regulation of this process, we applied an unbiased, quantitative, single-cell-resolution image analysis pipeline to analyze embryos lacking or exhibiting reduced levels of GATA6. We find that Gata6 mutants exhibit a complete absence of PrE and demonstrate that GATA6 levels regulate the timing and speed of lineage commitment within the ICM. Furthermore, we show that GATA6 is necessary for PrE specification by FGF signaling and propose a model where interactions between NANOG, GATA6, and the FGF/ERK pathway determine ICM cell fate. This study provides a framework for quantitative analyses of mammalian embryos and establishes GATA6 as a nodal point in the gene regulatory network driving ICM lineage specification.


Assuntos
Massa Celular Interna do Blastocisto/citologia , Endoderma/embriologia , Fator de Transcrição GATA6/metabolismo , Proteínas de Homeodomínio/biossíntese , Animais , Benzamidas/farmacologia , Diferenciação Celular , Linhagem da Célula , Difenilamina/análogos & derivados , Difenilamina/farmacologia , Técnicas de Cultura Embrionária , Embrião de Mamíferos/metabolismo , Endoderma/citologia , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator 4 de Crescimento de Fibroblastos/metabolismo , Fator de Transcrição GATA6/biossíntese , Fator de Transcrição GATA6/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas HMGB/biossíntese , Proteínas de Homeodomínio/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Knockout , Proteína Homeobox Nanog , Fatores de Transcrição SOXF/biossíntese
7.
Circ Res ; 115(2): 215-26, 2014 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-24755984

RESUMO

RATIONALE: The Notch pathway stabilizes sprouting angiogenesis by favoring stalk cells over tip cells at the vascular front. Because tip and stalk cells have different properties in morphology and function, their transcriptional regulation remains to be distinguished. Transcription factor Sox17 is specifically expressed in endothelial cells, but its expression and role at the vascular front remain largely unknown. OBJECTIVE: To specify the role of Sox17 and its relationship with the Notch pathway in sprouting angiogenesis. METHODS AND RESULTS: Endothelial-specific Sox17 deletion reduces sprouting angiogenesis in mouse embryonic and postnatal vascular development, whereas Sox17 overexpression increases it. Sox17 promotes endothelial migration by destabilizing endothelial junctions and rearranging cytoskeletal structure and upregulates expression of several genes preferentially expressed in tip cells. Interestingly, Sox17 expression is suppressed in stalk cells in which Notch signaling is relatively high. Notch activation by overexpressing Notch intracellular domain reduces Sox17 expression both in primary endothelial cells and in retinal angiogenesis, whereas Notch inhibition by delta-like ligand 4 (Dll4) blockade increases it. The Notch pathway regulates Sox17 expression mainly at the post-transcriptional level. Furthermore, endothelial Sox17 ablation rescues vascular network from excessive tip cell formation and hyperbranching under Notch inhibition in developmental and tumor angiogenesis. CONCLUSIONS: Our findings demonstrate that the Notch pathway restricts sprouting angiogenesis by reducing the expression of proangiogenic regulator Sox17.


Assuntos
Células Endoteliais/metabolismo , Proteínas HMGB/fisiologia , Neovascularização Patológica/fisiopatologia , Neovascularização Fisiológica/fisiologia , Receptores Notch/fisiologia , Fatores de Transcrição SOXF/fisiologia , Transdução de Sinais/fisiologia , Animais , Carcinoma Pulmonar de Lewis/irrigação sanguínea , Diferenciação Celular , Movimento Celular , Citoesqueleto/ultraestrutura , Embrião de Mamíferos/irrigação sanguínea , Células-Tronco Embrionárias , Regulação da Expressão Gênica , Proteínas HMGB/biossíntese , Proteínas HMGB/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Junções Intercelulares/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Morfogênese/genética , Estrutura Terciária de Proteína , RNA Interferente Pequeno/farmacologia , Receptor Notch1/genética , Receptor Notch1/fisiologia , Proteínas Recombinantes de Fusão , Vasos Retinianos/crescimento & desenvolvimento , Fatores de Transcrição SOXF/biossíntese , Fatores de Transcrição SOXF/genética , Organismos Livres de Patógenos Específicos , Transcrição Gênica
8.
Stem Cells Dev ; 17(4): 737-49, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18788933

RESUMO

Mouse embryonic fibroblasts (MEFs) have been extensively used as feeder cells to support the in vitro propagation of human embryonic stem cells (hESCs). However, owing to the risk of cross-contamination with animal or other unknown pathogens, the use of MEFs does not meet requirements for the clinical application of hESCs. Moreover, the actual role played by the feeders in the differentiation of hESCs is still unclear. In this study, human embryonic fibroblasts (HEFs) were used as feeder cells to support the establishment and undifferentiated growth of hESCs, and the capability of HEFs to induce the differentiation of definitive endoderm (DE) was evaluated. Three new hES cell lines were derived. These cell lines exhibited and maintained the common features of traditional hESCs after prolonged culture in vitro. Furthermore, DE differentiation of the newly established hES cell lines was performed using 100 ng/ml activin A, and the effects were compared among HEFs, MEFs, and feeder-free systems. On day 5 of induction, DE (SOX17(+)) cells appeared with comparable efficiency in both human and mouse feeder systems (85.0 +/- 8.9% and 78.7 +/- 3.4%, respectively). These levels were considerably superior to that obtained in the feeder-free system (22.7 +/- 5.6%). The SOX17(+) cells tended to differentiate into an endodermal lineage in vivo and could be further induced into glucagon and C-peptide double positive islet-like clusters in vitro. Our studies suggest that, in terms of therapeutic application, HEFs can be an effective substitute for MEFs for sustaining the derivation and DE differentiation of hESCs.


Assuntos
Diferenciação Celular/fisiologia , Embrião de Mamíferos/citologia , Células-Tronco Embrionárias/citologia , Fibroblastos/citologia , Ativinas/farmacologia , Animais , Peptídeo C/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Proteínas de Ligação a DNA/biossíntese , Embrião de Mamíferos/metabolismo , Células-Tronco Embrionárias/metabolismo , Endoderma/citologia , Endoderma/metabolismo , Fibroblastos/metabolismo , Glucagon/farmacologia , Proteínas HMGB/biossíntese , Proteínas de Grupo de Alta Mobilidade/biossíntese , Humanos , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Fatores de Transcrição SOXF , Fatores de Transcrição/biossíntese
9.
Stem Cells ; 26(4): 903-11, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18238855

RESUMO

Previous studies have demonstrated that the transcription factor Sox2 is essential during the early stages of development. Furthermore, decreasing the expression of Sox2 severely interferes with the self-renewal and pluripotency of embryonic stem (ES) cells. Other studies have shown that Sox2, in conjunction with the transcription factor Oct-3/4, stimulates its own transcription as well as the expression of a growing list of genes (Sox2:Oct-3/4 target genes) that require the cooperative action of Sox2 and Oct-3/4. Remarkably, recent studies have shown that overexpression of Sox2 decreases expression of its own gene, as well as four other Sox2:Oct-3/4 target genes (Oct-3/4, Nanog, Fgf-4, and Utf1). This finding led to the prediction that overexpression of Sox2 in ES cells would trigger their differentiation. In the current study, we initially engineered mouse ES cells for inducible overexpression of Sox2. Using this model system, we demonstrate that small increases (twofold or less) in Sox2 protein trigger the differentiation of ES cells into cells that exhibit markers for a wide range of differentiated cell types, including neuroectoderm, mesoderm, and trophectoderm but not endoderm. We also demonstrate that elevating the levels of Sox2 quickly downregulates several developmentally regulated genes, including Nanog, and a newly identified Sox2:Oct-3/4 target gene, Lefty1. Together, these data argue that the self-renewal of ES cells requires that Sox2 levels be maintained within narrow limits. Thus, Sox2 appears to function as a molecular rheostat that controls the expression of a critical set of embryonic genes, as well as the self-renewal and differentiation of ES cells.


Assuntos
Diferenciação Celular/fisiologia , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Proteínas HMGB/biossíntese , Proteínas HMGB/fisiologia , Fatores de Transcrição/biossíntese , Fatores de Transcrição/fisiologia , Animais , Diferenciação Celular/genética , Linhagem Celular , Proteínas de Ligação a DNA/genética , Evolução Molecular Direcionada/métodos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas HMGB/genética , Camundongos , Fatores de Transcrição SOXB1 , Fatores de Transcrição/genética
10.
J Neurosci Res ; 86(8): 1680-93, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18293417

RESUMO

SOX2 is a key neurodevelopmental gene involved in maintaining the pluripotency of stem cells and proliferation of neural progenitors and astroglia. Two evolutionally conserved enhancers, SRR1 and SRR2, are involved in controlling SOX2 expression during neurodevelopment; however, the molecular mechanisms regulating their activity are not known. We have examined DNA methylation and histone H3 acetylation at both enhancers in NT2-D1 progenitors, neurons and astrocytes, to establish the role of epigenetic mechanisms in cell-type-specific SOX2 expression. This study showed that 1) unmethylated DNA and acetylated histones at both enhancers correlated with a high level of SOX2 expression in proliferating neural progenitors and 2) reversible modifications of the SRR1 element were observed during gene reexpression in astrocytes, whereas permanent epigenetic marks on the SRR2 enhancer were seen in neurons where the gene was silenced. Taken together, these results are clear illustrations of cell-type-specific epigenomes and suggest mechanisms by which they may be created and maintained.


Assuntos
Proteínas de Ligação ao Cálcio/fisiologia , Diferenciação Celular/fisiologia , Proteínas de Ligação a DNA/biossíntese , Elementos Facilitadores Genéticos/fisiologia , Epigênese Genética/fisiologia , Proteínas HMGB/biossíntese , Glicoproteínas de Membrana/fisiologia , Neurônios/citologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Receptores de Peptídeos/fisiologia , Fatores de Transcrição/biossíntese , Acetilação , Astrócitos/citologia , Astrócitos/metabolismo , Sequência de Bases , Proteínas de Ligação ao Cálcio/genética , Células Cultivadas , Metilação de DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas HMGB/genética , Proteínas HMGB/metabolismo , Humanos , Glicoproteínas de Membrana/genética , Dados de Sequência Molecular , Neurônios/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores de Peptídeos/genética , Fatores de Transcrição SOXB1 , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
11.
PLoS Biol ; 6(1): e2, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18184035

RESUMO

In vertebrate embryos, the earliest definitive marker for the neural plate, which will give rise to the entire central nervous system, is the transcription factor Sox2. Although some of the extracellular signals that regulate neural plate fate have been identified, we know very little about the mechanisms controlling Sox2 expression and thus neural plate identity. Here, we use electroporation for gain- and loss-of-function in the chick embryo, in combination with bimolecular fluorescence complementation, two-hybrid screens, chromatin immunoprecipitation, and reporter assays to study protein interactions that regulate expression of N2, the earliest enhancer of Sox2 to be activated and which directs expression to the largest part of the neural plate. We show that interactions between three coiled-coil domain proteins (ERNI, Geminin, and BERT), the heterochromatin proteins HP1alpha and HP1gamma acting as repressors, and the chromatin-remodeling enzyme Brm acting as activator control the N2 enhancer. We propose that this mechanism regulates the timing of Sox2 expression as part of the process of establishing neural plate identity.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Proteínas HMGB/biossíntese , Placa Neural/metabolismo , Fatores de Transcrição/biossíntese , Sequência de Aminoácidos , Animais , Proteínas Aviárias/metabolismo , Proteínas de Ciclo Celular/metabolismo , Embrião de Galinha , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/genética , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica no Desenvolvimento , Proteínas HMGB/genética , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/metabolismo , Placa Neural/embriologia , Ligação Proteica , Fatores de Transcrição SOXB1 , Fatores de Transcrição/genética
12.
Am J Surg Pathol ; 32(1): 103-12, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18162777

RESUMO

Sox2 is a key transcription factor that maintains the proliferation of neuroglial stem cells and inhibits neuronal fate commitment. Moreover, it was recently found that brain tumors contain stem cells that resemble normal neuroglial stem cells in many respects. This study was undertaken to describe Sox2 expression in various brain tumors, and to determine whether Sox2 expression is a universal feature of brain tumors, or whether its expression is limited to a specific lineage of brain tumors. Sox2 immunohistochemistry was performed on 194 brain tumor tissues of various kinds. Fetal and adult normal brain tissues obtained by autopsy and brain tissues of epilepsy patients with cortical dysplasia were used as controls. Semiquantitative reverse transcription polymerase chain reaction was used to confirm the immunohistochemical results. Double immunofluorescence was performed to characterize the lineage of Sox2-positive cells. Sox2 was found to be expressed in various glial tumors, including those with astroglial, oligodendroglial, and ependymal lineages, and in the glial components of mixed neuroglial tumors, regardless of pathologic grade. In brain tumors of embryonal origin, supratentorial primitive neuroectodermal tumors showed robust Sox2 expression, whereas medulloblastomas and pineoblastomas did not. The majority of Sox2-positive tumor cells coexpressed glial fibrillary acidic protein, and most Sox2-negative cells in medulloblastomas and pineoblastomas showed neuronal differentiation. This study suggest that Sox2 may be a tumor marker of glial lineages rather than a universal brain tumor stem cell marker, because its expression pattern was found to correspond to differentiation pathways. On the other hand, the aberrant coexpressions of Sox2 and of a neuronal marker were widely observed in glioblastomas, which reflects a disorganized differentiation pattern that characterizes highly malignant tumors.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias Encefálicas/metabolismo , Proteínas HMGB/biossíntese , Neuroglia/citologia , Neuroglia/metabolismo , Fatores de Transcrição/biossíntese , Encéfalo/citologia , Encéfalo/embriologia , Encéfalo/metabolismo , Neoplasias Encefálicas/patologia , Diferenciação Celular , Linhagem da Célula , Imunofluorescência , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Imuno-Histoquímica , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOXB1
13.
Dev Dyn ; 236(9): 2702-7, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17685482

RESUMO

Sox5 is a member of the SoxD group of HMG-box transcription factors that, during the early stages of development, promotes neural crest generation. However, little is known about Sox5 function in neural crest derivatives such as the peripheral sensory nervous system. We have analysed the embryonic expression of Sox5 during chick cranial ganglia development, from the stages of ganglia condensation to those of differentiation. During this period, Sox5 expression is maintained in the crest-derived satellite glial cells in all the cranial ganglia. In contrast, Sox5 is only transiently expressed in a subpopulation of differentiating neurons of both neural crest and placode origin. This detailed analysis provides a good base to dissect the possible role of Sox5 in neural cell fate determination by future functional approaches.


Assuntos
Encéfalo/embriologia , Gânglios/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas HMGB/biossíntese , Proteínas HMGB/genética , Neuroglia/metabolismo , Animais , Diferenciação Celular , Linhagem da Célula , Embrião de Galinha , Proteínas de Ligação a DNA/biossíntese , Perfilação da Expressão Gênica , Proteínas de Grupo de Alta Mobilidade/biossíntese , Imuno-Histoquímica , Crista Neural/embriologia , Fatores de Transcrição SOXE , Fatores de Transcrição/biossíntese , Gânglio Trigeminal/embriologia
14.
Biol Reprod ; 77(3): 577-88, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17494917

RESUMO

The amnion is the inner of two membranes surrounding the fetus. That it arises from embryonic epiblast cells prior to gastrulation suggests that it may retain a reservoir of stem cells throughout pregnancy. We found that human amniotic epithelial cells (hAECs) harvested from term-delivered fetal membranes express mRNA and proteins present in human embryonic stem cells (hESCs), including POU domain, class 5, transcription factor 1; Nanog homeobox; SRY-box 2; and stage-specific embryonic antigen-4. In keeping with possible stem cell-like activity, hAECs were also clonogenic, and primary hAEC cultures could be induced to differentiate into cardiomyocytic, myocytic, osteocytic, adipocytic (mesodermal), pancreatic, hepatic (endodermal), neural, and astrocytic (neuroectodermal) cells in vitro, as defined by phenotypic, mRNA expression, immunocytochemical, and/or ultrastructural characteristics. However, unlike hESCs, hAECs did not form teratomas upon transplantation into severe combined immunodeficiency mice testes. Last, using flow cytometry we have shown that only a very small proportion of primary hAECs contain class IA and class II human leukocyte antigens (HLAs), consistent with a low risk of tissue rejection. However, following differentiation into hepatic and pancreatic lineages, significant proportions of cells contained class IA, but not class II, HLAs. These observations suggest that the term amnion, an abundant and easily accessible tissue, may be a useful source of multipotent stem cells that possess a degree of immune privilege.


Assuntos
Âmnio/fisiologia , Diferenciação Celular/fisiologia , Células-Tronco Fetais/citologia , Células-Tronco Multipotentes/citologia , Animais , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Feminino , Células-Tronco Fetais/metabolismo , Citometria de Fluxo , Glicoesfingolipídeos/biossíntese , Glicoesfingolipídeos/genética , Proteínas HMGB/biossíntese , Proteínas HMGB/genética , Antígenos de Histocompatibilidade Classe I/biossíntese , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe II/biossíntese , Antígenos de Histocompatibilidade Classe II/genética , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos SCID , Células-Tronco Multipotentes/metabolismo , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/biossíntese , Fator 3 de Transcrição de Octâmero/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOXB1 , Antígenos Embrionários Estágio-Específicos , Transplante de Células-Tronco , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
15.
Mod Pathol ; 20(7): 742-8, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17464316

RESUMO

Sox2 is a transcription factor that plays a critical role in the maintenance of the self-renewal capability of neural stem cells. This study was undertaken to investigate the expression pattern of Sox2 in mature and immature teratomas of the central nervous system. Sox2 immunohistochemistry was performed in 14 cases of central nervous system teratoma: five mature, five immature teratomas, and four mixed germ cell tumors with a prominent teratoma component. Fetal brain tissue was used as a normal control. Immunofluorescence with double labeling of stem cells and neuroglial cell markers was used for phenotyping of Sox2-positive cells. In all cases of immature teratomas, positive reactivity to Sox2 was observed in primitive neuroepithelial tissues. Sox2 was not expressed in mature tissues, except in some cuboidal or columnar epithelium of endodermal origin. In mature teratomas, Sox2 expression was limited to some endodermal epithelium in two cases, and no Sox2 expression was observed in the other three cases. The majority of Sox2-positive neuroepithelial cells also expressed neural stem cell markers, nestin and vimentin. Sox2 and neuronal and oligodendroglial markers were expressed in a mutually exclusive manner. However, mature astroglial cells coexpressed Sox2 and GFAP. In fetal brain, Sox2 was mainly expressed in ventricular and subventricular zones. Since Sox2 is strongly expressed in the primitive neuroepithelial tissues of central nervous system immature teratomas, it may be a useful biomarker for the diagnosis and quantitative grading of central nervous system immature teratomas.


Assuntos
Neoplasias do Sistema Nervoso Central/patologia , Proteínas HMGB/biossíntese , Teratoma/patologia , Fatores de Transcrição/biossíntese , Adolescente , Adulto , Neoplasias do Sistema Nervoso Central/metabolismo , Criança , Pré-Escolar , Feminino , Imunofluorescência , Proteína Glial Fibrilar Ácida/análise , Humanos , Imuno-Histoquímica , Lactente , Masculino , Microscopia Confocal , Fatores de Transcrição SOXB1 , Teratoma/metabolismo
16.
Dev Neurobiol ; 67(5): 637-54, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17443814

RESUMO

Mechanosensory hair cells are essential for audition in vertebrates, and in many species, have the capacity for regeneration when damaged. Regeneration is robust in the fish lateral line system as new hair cells can reappear after damage induced by waterborne aminoglycoside antibiotics, platinum-based drugs, and heavy metals. Here, we characterize the loss and reappearance of lateral line hair cells induced in zebrafish larvae treated with copper sulfate using diverse molecular markers. Transgenic fish that express green fluorescent protein in different cell types in the lateral line system have allowed us to follow the regeneration of hair cells after different damage protocols. We show that conditions that damage only differentiated hair cells lead to reappearance of new hair cells within 24 h from nondividing precursors, whereas harsher conditions are followed by a longer recovery period that is accompanied by extensive cell division. In order to characterize the cell population that gives rise to new hair cells, we describe the expression of a neural stem cell marker in neuromasts. The zebrafish sox2 gene is strongly expressed in neuromast progenitor cells, including those of the migrating lateral line primordium, the accessory cells that underlie the hair cells in neuromasts, and in interneuromastic cells that give rise to new neuromasts. Moreover, we find that most of the cells that proliferate within the neuromast during regeneration express this marker. Thus, our results describe the dynamics of hair cell regeneration in zebrafish and suggest the existence of at least two mechanisms for recovery of these cells in neuromasts.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Proteínas HMGB/biossíntese , Células Ciliadas Auditivas/fisiologia , Sistema da Linha Lateral/fisiologia , Neurônios/fisiologia , Células-Tronco/fisiologia , Fatores de Transcrição/biossíntese , Animais , Animais Geneticamente Modificados , Antimetabólitos , Bromodesoxiuridina , Proliferação de Células/efeitos dos fármacos , Cobre/toxicidade , Proteínas de Ligação a DNA/genética , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Proteínas HMGB/genética , Células Ciliadas Auditivas/efeitos dos fármacos , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Hibridização In Situ , Mitose/fisiologia , Regeneração Nervosa/fisiologia , Fatores de Transcrição SOXB1 , Células-Tronco/efeitos dos fármacos , Fatores de Transcrição/genética , Peixe-Zebra
17.
Dev Biol ; 299(2): 438-54, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16959242

RESUMO

The zebrafish caudal fin provides a simple model to study molecular mechanisms of dermal bone regeneration. We previously showed that misexpression of Bone morphogenetic protein 2b (Bmp2b) induces ectopic bone formation within the regenerate. Here we show that in addition to bmp2b and bmp4 another family member, bmp6, is involved in fin regeneration. We further investigated the function of BMP signaling by ectopically expressing the BMP signaling inhibitor Chordin which caused: (1) inhibition of regenerate outgrowth due to a decrease of blastema cell proliferation and downregulation of msxb and msxC expression and (2) reduced bone matrix deposition resulting from a defect in the maturation and function of bone-secreting cells. We then identified targets of BMP signaling involved in regeneration of the bone of the fin rays. runx2a/b and their target col10a1 were downregulated following BMP signaling inhibition. Unexpectedly, the sox9a/b transcription factors responsible for chondrocyte differentiation were detected in the non-cartilaginous fin rays, sox9a and sox9b were not only differentially expressed but also differentially regulated since sox9a, but not sox9b, was downregulated in the absence of BMP signaling. Finally, this analysis revealed the surprising finding of the expression, in the fin regenerate, of several factors which are normally the signatures of chondrogenic elements during endochondral bone formation although fin rays form through dermal ossification, without a cartilage intermediate.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Diferenciação Celular , Osteoblastos/citologia , Regeneração , Peixe-Zebra/anatomia & histologia , Peixe-Zebra/fisiologia , Animais , Proteína Morfogenética Óssea 2 , Proteína Morfogenética Óssea 4 , Proteína Morfogenética Óssea 6 , Proteínas Morfogenéticas Ósseas/metabolismo , Proliferação de Células , Condrócitos/citologia , Regulação para Baixo , Glicoproteínas/biossíntese , Proteínas HMGB/biossíntese , Proteínas de Homeodomínio/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Osteogênese , Fatores de Transcrição SOX9 , Transdução de Sinais , Fatores de Transcrição/metabolismo , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/biossíntese , Proteínas de Peixe-Zebra/metabolismo
18.
Clin Genet ; 69(6): 459-70, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16712695

RESUMO

Eye development initiates as an evagination of the early neural plate, before the closure of the neural tube. Structural malformations of the eye such as anophthalmia and microphthalmia arise very early in development. It is not surprising therefore that three of the genes currently identified to play a significant role in these developmental eye anomalies are also major players in brain development and regionalization. However, as has been emerging for a high proportion of transcriptional regulators studied, these genes have evolved to play multiple roles throughout development, and perhaps even in adult tissue maintenance. This complex spatiotemporal expression pattern requires elaborate regulatory systems which we are beginning to unravel. A major component of these complex regulatory networks is a series of cis-acting elements, highly conserved through evolution, which spread large distances from the coding region of each gene. We describe how cross regulation for PAX6, SOX2 and perhaps OTX2 has now been uncovered, pointing to the mechanisms that can fine-tune the expression of such essential developmental components. These interactions also help us understand why there is significant phenotypic overlap between mutations at these three loci.


Assuntos
Proteínas de Ligação a DNA/genética , Anormalidades do Olho/embriologia , Anormalidades do Olho/genética , Proteínas do Olho/genética , Proteínas HMGB/genética , Proteínas de Homeodomínio/genética , Fatores de Transcrição Otx/genética , Fatores de Transcrição Box Pareados/genética , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Animais , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/fisiologia , Proteínas do Olho/biossíntese , Proteínas do Olho/fisiologia , Proteínas HMGB/biossíntese , Proteínas HMGB/fisiologia , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/fisiologia , Humanos , Fatores de Transcrição Otx/biossíntese , Fatores de Transcrição Otx/fisiologia , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/biossíntese , Fatores de Transcrição Box Pareados/fisiologia , Proteínas Repressoras/biossíntese , Proteínas Repressoras/fisiologia , Fatores de Transcrição SOXB1 , Fatores de Transcrição/biossíntese , Fatores de Transcrição/fisiologia
19.
Cancer Res ; 66(2): 763-9, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16424007

RESUMO

Identification of biomarkers could lead to the development of effective screening tests for colorectal cancer. A previous study from our laboratory showed specific alterations of nuclear structure in colon cancer. In an effort to characterize these biomarkers, protein spots were selected from separations made by two-dimensional gel electrophoresis, which were analyzed by mass spectrometry. The sequences obtained from the isolated spots revealed that they have close similarity to creatine kinase B (CKB) isoforms, heterogeneous nuclear ribonucleoprotein F (hnRNP F) and high mobility group box 1 protein (HMGB1) isoforms. To determine the expression of these proteins in colon cancer, expression was studied in 9 tumor and matched adjacent normal pairs, 5 donor colons, 16 polyps, 4 metastatic liver lesions and matched adjacent normal pairs, and 3 liver donors. CKB and hnRNP F were expressed in 78% and 89% of colon tumors, respectively. hnRNP F had a higher frequency of expression than CKB in premalignant polyps. With the establishment of differential expression of the proteins in colon cancer, their subcellular localization was analyzed. The subcellular fractions studied both showed high protein levels of hnRNP F in colon tumors compared with normal colon tissues. Surprisingly, subcellular levels of CKB were decreased in colon tumors, suggesting that the observed high CKB levels in nuclear matrix extracts are caused by the enhanced localization of CKB to the nuclear matrix during colon tumorigenesis. These results suggest an involvement of hnRNP F and CKB in colorectal cancer. Additionally, they suggest that hnRNP F is a potential marker for colorectal cancer progression.


Assuntos
Neoplasias do Colo/genética , Creatina Quinase Forma BB/biossíntese , Perfilação da Expressão Gênica , Proteínas HMGB/biossíntese , Ribonucleoproteínas Nucleares Heterogêneas Grupo F-H/biossíntese , Neoplasias Hepáticas/secundário , Adenocarcinoma/genética , Adenocarcinoma/patologia , Adulto , Idoso , Sequência de Aminoácidos , Biomarcadores Tumorais/análise , Estudos de Casos e Controles , Transformação Celular Neoplásica , Neoplasias do Colo/patologia , Pólipos do Colo/genética , Pólipos do Colo/patologia , Creatina Quinase Forma BB/análise , Creatina Quinase Forma BB/genética , Eletroforese em Gel Bidimensional , Feminino , Proteínas HMGB/análise , Proteínas HMGB/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo F-H/análise , Ribonucleoproteínas Nucleares Heterogêneas Grupo F-H/genética , Humanos , Neoplasias Hepáticas/genética , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular
20.
Aging Cell ; 4(4): 197-207, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16026334

RESUMO

The identification of neural stem cells (NSCs) in situ has been prevented by the inability to identify a marker consistently expressed in all adult NSCs and is thus generally accomplished using the in vitro neurosphere-forming assay. The high-mobility group transcription factor Sox2 is expressed in embryonic neural epithelial stem cells; because these cells are thought to give rise to the adult NSC population, we hypothesized that Sox2 may continue to be expressed in adult NSCs. Using Sox2:EGFP transgenic mice, we show that Sox2 is expressed in neurogenic regions along the rostral-caudal axis of the central nervous system throughout life. Furthermore, all neurospheres derived from these neurogenic regions express Sox2, suggesting that Sox2 is indeed expressed in adult NSCs. We demonstrate that NSCs are heterogeneous within the adult brain, with differing capacities for cell production. In vitro, all neurospheres express Sox2, but the expression of markers common to early progenitor cells within individual neurospheres varies; this heterogeneity of NSCs is mirrored in vivo. For example, both glial fibrillary acidic protein and NG2 are expressed within individual neurospheres, but their expression is mutually exclusive; likewise, these two markers show distinct staining patterns within the Sox2+ regions of the brain's neurogenic regions. Thus, we propose that the expression of Sox2 is a unifying characteristic of NSCs in the adult brain, but that not all NSCs maintain the ability to form all neural cell types in vivo.


Assuntos
Encéfalo/metabolismo , Proteínas de Ligação a DNA/biossíntese , Proteínas HMGB/biossíntese , Neurônios/metabolismo , Células-Tronco/metabolismo , Fatores de Transcrição/biossíntese , Fatores Etários , Animais , Encéfalo/citologia , Diferenciação Celular/fisiologia , Células Cultivadas , DNA Complementar/metabolismo , Proteínas de Ligação a DNA/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Proteínas HMGB/genética , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/citologia , Gravidez , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOXB1 , Células-Tronco/citologia , Telomerase/metabolismo , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA