Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(31): e2123467119, 2022 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-35881788

RESUMO

Capicua (Cic) proteins are conserved HMG-box transcriptional repressors that control receptor tyrosine kinase (RTK) signaling responses and are implicated in human neurological syndromes and cancer. While Cic is known to exist as short (Cic-S) and long (Cic-L) isoforms with identical HMG-box and associated core regions but distinct N termini, most previous studies have focused on Cic-S, leaving the function of Cic-L unexplored. Here we show that Cic-L acts in two capacities during Drosophila oogenesis: 1) as a canonical sensor of RTK signaling in somatic follicle cells, and 2) as a regulator of postmitotic growth in germline nurse cells. In these latter cells, Cic-L behaves as a temporal signal that terminates endoreplicative growth before they dump their contents into the oocyte. We show that Cic-L is necessary and sufficient for nurse cell endoreplication arrest and induces both stabilization of CycE and down-regulation of Myc. Surprisingly, this function depends mainly on the Cic-L-specific N-terminal module, which is capable of acting independently of the Cic HMG-box-containing core. Mirroring these observations, basal metazoans possess truncated Cic-like proteins composed only of Cic-L N-terminal sequences, suggesting that this module plays unique, ancient roles unrelated to the canonical function of Cic.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster , Proteínas HMGB , Oogênese , Proteínas Repressoras , Animais , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/fisiologia , Proteínas HMGB/genética , Proteínas HMGB/fisiologia , Oogênese/genética , Receptores Proteína Tirosina Quinases/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/fisiologia
2.
Cell Immunol ; 352: 104085, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32201004

RESUMO

Asthma is a disease of the respiratory system that is commonly considered a T-helper 2 (Th2) cell-associated inflammatory disease. Group 2 innate lymphoid cells (ILC2s) promote the inflammatory responses in asthma by secreting type 2 cytokines. Interleukin (IL)-9 also serves as a promoting factor in asthma and it is well known that ILC2s have an autocrine effect of IL-9 to sustain their survival and proliferation. However, the specific role of ILC2-derived IL-9 in asthma remains unclear. HMGB1 (High-Mobility Group Box-1) is a nuclear protein, and Previous studies have shown that HMGB1 can regulate the differentiation of T-helper cells and participate in the development of asthma. But whether HMGB1 can regulate the innate lymphocytes in the pathological process of asthma is unknown. In this study we have shown increased presence of HMGB1 protein in the lung of mice with asthma, which was associated with increased secretion of IL-9 by ILC2s. This led to the activation of dendritic cells (DCs) that can accelerate the differentiation of Th2 cells and worsen the severity of asthma. Taken together, our study provides a complementary understanding of the asthma development and highlights a novel inflammatory pathway in the pathogenesis of asthma.


Assuntos
Asma/metabolismo , Proteína HMGB1/metabolismo , Linfócitos/imunologia , Animais , Asma/fisiopatologia , Diferenciação Celular/efeitos dos fármacos , Citocinas/metabolismo , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Feminino , Proteínas HMGB/metabolismo , Proteínas HMGB/fisiologia , Proteína HMGB1/fisiologia , Imunidade Inata/efeitos dos fármacos , Interleucina-9/imunologia , Interleucina-9/metabolismo , Pulmão/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Células Th2/imunologia
3.
Brain Behav Immun ; 82: 167-177, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31430517

RESUMO

High-fat high-sugar diet-induced obesity can lead to hippocampal inflammation and cognitive deficits, but the detailed underlying mechanism is still not clear. We aim to investigate the role of HMGB1 in hippocampal inflammatory responses and cognitive impairment in high-fat high-fructose diet (HFHFD)-induced obesity. Rats were fed with a normal control diet or an HFHFD diet for 14 weeks. In the last 6 weeks on the diets, the rats were treated with control, or an HMGB1 inhibitor glycyrrhizin, or an anti-HMGB1 neutralizing monoclonal antibody (mAb). Obesity was induced in the HFHFD-fed rats, which had higher body weight, epididymal white adipose tissue (EWAT) weight and caloric efficiency, and lower brain/body weight ratio, glucose tolerance and insulin sensitivity than the ones on normal diets. In the HFHFD-induced obese rats, the HMGB1 levels in plasma and hippocampus were increased, and the nucleus-to-cytoplasm translocation of HMGB1 was promoted. The hippocampal inflammatory responses were enhanced in the HFHFD-induced obesity, including the activation of TLR4 and NF-κB, the production of IL-1ß, TNF-α and IL-6, as well as the activation of microglia and astrocytes. In addition, the hippocampal cell apoptosis and cognitive impairment were observed in the HFHFD-fed rats. The treatment with glycyrrhizin or HMGB1 mAb successfully decreased the HMGB1 levels in plasma and hippocampus, and prevented the HMGB1 translocation from the nucleus to cytoplasm. Inhibiting HMGB1 by glycyrrhizin or HMGB1 mAb suppressed the hippocampal inflammatory, alleviated the apoptosis and ameliorated the cognitive impairment in HFHFD-fed rats. These findings indicate that HMGB1 mediates the hippocampal inflammation and contributes to the cognitive deficits in HFHFD-induced obesity. Therefore, inhibition of HMGB1 may have beneficial effect in protecting against hippocampal inflammation and cognitive deficits in dietary obesity.


Assuntos
Disfunção Cognitiva/etiologia , Proteína HMGB1/metabolismo , Obesidade/metabolismo , Animais , Apoptose/efeitos dos fármacos , Cognição/fisiologia , Disfunção Cognitiva/metabolismo , Dieta Hiperlipídica/efeitos adversos , Ácido Glicirrízico/farmacologia , Proteínas HMGB/metabolismo , Proteínas HMGB/fisiologia , Proteína HMGB1/fisiologia , Hipocampo/metabolismo , Inflamação , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Masculino , Microglia/metabolismo , NF-kappa B/metabolismo , Obesidade/fisiopatologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
4.
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
5.
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
6.
Int J Parasitol ; 46(4): 253-62, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26820302

RESUMO

The Schistosoma mansoni High Mobility Group Box (HMGB) proteins SmHMGB1, SmHMGB2 and SmHMGB3 share highly conserved HMG box DNA binding domains but have significantly different C-terminal acidic tails. Here, we used three full-length and tailless forms of the S. mansoni HMGB proteins to examine the functional roles of their acidic tails. DNA binding assays revealed that the different lengths of the acidic tails among the three SmHMGB proteins significantly and distinctively influenced their DNA transactions. Spectroscopic analyses indicated that the longest acidic tail of SmHMGB3 contributes to the structural stabilisation of this protein. Using immunohistochemical analysis, we showed distinct patterns of SmHMGB1, SmHMGB2 and SmHMGB3 expression in different tissues of adult worms. RNA interference approaches indicated a role for SmHMGB2 and SmHMGB3 in the reproductive system of female worms, whereas for SmHMGB1 no clear phenotype was observed. Schistosome HMGB proteins can be phosphorylated, acetylated and methylated. Importantly, the acetylation and methylation of schistosome HMGBs were greatly enhanced upon removal of the acidic tail. These data support the notion that the C-terminal acidic tails dictate the differences in the structure, expression and function of schistosome HMGB proteins.


Assuntos
Proteínas HMGB/química , Schistosoma mansoni/química , Sequência de Aminoácidos , Animais , Dicroísmo Circular , DNA de Helmintos/metabolismo , Feminino , Proteínas HMGB/genética , Proteínas HMGB/fisiologia , Imuno-Histoquímica , Masculino , Especificidade de Órgãos , Domínios e Motivos de Interação entre Proteínas/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Interferência de RNA , Schistosoma mansoni/genética
7.
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
8.
Circulation ; 131(11): 995-1005, 2015 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-25596186

RESUMO

BACKGROUND: Intracranial aneurysm (IA) is a common vascular disorder that frequently leads to fatal vascular rupture. Although various acquired risk factors associated with IA have been identified, the hereditary basis of IA remains poorly understood. As a result, genetically modified animals accurately modeling IA and related pathogenesis have been lacking, and subsequent drug development has been delayed. METHODS AND RESULTS: The transcription factor Sox17 is robustly expressed in endothelial cells of normal intracerebral arteries. The combination of Sox17 deficiency and angiotensin II infusion in mice induces vascular abnormalities closely resembling the cardinal features of IA such as luminal dilation, wall thinning, tortuosity, and subarachnoid hemorrhages. This combination impairs junctional assembly, cell-matrix adhesion, regeneration capacity, and paracrine secretion in endothelial cells of intracerebral arteries, highlighting key endothelial dysfunctions that lead to IA pathogenesis. Moreover, human IA samples showed reduced Sox17 expression and impaired endothelial integrity, further strengthening the applicability of this animal model to clinical settings. CONCLUSIONS: Our findings demonstrate that Sox17 deficiency in mouse can induce IA under hypertensive conditions, suggesting Sox17 deficiency as a potential genetic factor for IA formation. The Sox17-deficient mouse model provides a novel platform to develop therapeutics for incurable IA.


Assuntos
Endotélio Vascular/patologia , Proteínas HMGB/deficiência , Aneurisma Intracraniano/genética , Fatores de Transcrição SOXF/deficiência , Fatores de Transcrição SOXF/fisiologia , Adulto , Idoso , Angiotensina II/toxicidade , Animais , Aorta/patologia , Células Cultivadas , Artérias Cerebrais/química , Artérias Cerebrais/patologia , Proteínas Inibidoras de Quinase Dependente de Ciclina/biossíntese , Proteínas Inibidoras de Quinase Dependente de Ciclina/genética , Dilatação Patológica/genética , Dilatação Patológica/patologia , Modelos Animais de Doenças , Endotélio Vascular/metabolismo , Feminino , Proteínas HMGB/genética , Proteínas HMGB/fisiologia , Humanos , Hipertensão/complicações , Aneurisma Intracraniano/etiologia , Aneurisma Intracraniano/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Miócitos de Músculo Liso/química , Comunicação Parácrina , Interferência de RNA , Fatores de Transcrição SOXF/análise , Fatores de Transcrição SOXF/genética , Organismos Livres de Patógenos Específicos , Hemorragia Subaracnóidea/etiologia , Transcrição Gênica , Regulação para Cima , Veias/química
9.
Nat Commun ; 5: 5434, 2014 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-25399555

RESUMO

Sex determination in animals and fungi is regulated by specific sex-determining genes. The Aspergillus nidulans mating type gene matA and the human SRY (Sex-Determining Region Y) encode proteins containing a single HMG (high-mobility group) domain. Analysis of the amino-acid sequence of MatA and SRY transcription factors revealed significant structural similarity. The human SRY protein is able to functionally replace MatA and drives the sexual cycle in the fungus A. nidulans. Functional studies indicate that SRY drives early fruiting body development, and hybrid MatA protein carrying the SRY HMG box is fully capable of driving both early and late stages of sexual development, including gametogenesis. Our data suggest that SRY and MatA are both structurally and functionally related and conserved in regulating sexual processes. The fundamental mechanisms driving evolution of the genetic pathways underlying sex determination, sex chromosomes and sexual reproduction in eukaryotes appear similar.


Assuntos
Aspergillus nidulans/fisiologia , Proteínas Fúngicas/fisiologia , Genes Fúngicos Tipo Acasalamento/fisiologia , Processos de Determinação Sexual/fisiologia , Proteína da Região Y Determinante do Sexo/fisiologia , Sequência de Aminoácidos , Aspergillus nidulans/genética , Carpóforos/genética , Carpóforos/crescimento & desenvolvimento , Carpóforos/fisiologia , Proteínas Fúngicas/genética , Genes Fúngicos Tipo Acasalamento/genética , Proteínas HMGB/genética , Proteínas HMGB/fisiologia , Humanos , Dados de Sequência Molecular , Homologia de Sequência , Processos de Determinação Sexual/genética , Proteína da Região Y Determinante do Sexo/genética
11.
Biochim Biophys Acta ; 1839(9): 764-72, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24972368

RESUMO

The regulation of gene expression at the level of transcription involves the concerted action of several proteins and protein complexes committed to dynamically alter the surrounding chromatin environment of a gene being activated or repressed. ATP-dependent chromatin remodeling complexes are key factors in chromatin remodeling, and the SWI/SNF complex is the founding member. While many studies have linked the action of these complexes to specific transcriptional regulation of a large number of genes and much is known about their catalytic activity, less is known about the nuclear elements that can enhance or modulate their activity. A number of studies have found that certain High Mobility Group (HMG) proteins are able to stimulate ATP-dependent chromatin remodeling activity, but their influence on the different biochemical outcomes of this activity is still unknown. In this work we studied the influence of the yeast Nhp6A, Nhp6B and Hmo1 proteins (HMGB family members) on different biochemical outcomes of yeast SWI/SNF remodeling activity. We found that all these HMG proteins stimulate the sliding activity of ySWI/SNF, while transient exposure of nucleosomal DNA and octamer transfer catalyzed by this complex are only stimulated by Hmo1. Consistently, only Hmo1 stimulates SWI/SNF binding to the nucleosome. Additionally, the sliding activity of another chromatin remodeling complex, ISW1a, is only stimulated by Hmo1. Further analyses show that these differential stimulatory effects of Hmo1 are dependent on the presence of its C-terminal tail, which contains a stretch of acidic and basic residues.


Assuntos
Montagem e Desmontagem da Cromatina , Proteínas Cromossômicas não Histona/fisiologia , Proteínas Fúngicas/fisiologia , Proteínas HMGB/fisiologia , Nucleossomos/fisiologia , Saccharomyces cerevisiae/metabolismo , Proteínas de Ligação a DNA/fisiologia , Proteínas HMGN/fisiologia , Proteínas de Grupo de Alta Mobilidade/fisiologia , Ligação Proteica , Proteínas de Saccharomyces cerevisiae/fisiologia
12.
Neurochem Int ; 75: 32-8, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24893328

RESUMO

To clarify the mechanism of high-mobility group box (HMGB) 1-induced brain edema formation, this study focused on the effect of HMGB1 on aquaporin (AQP) 4, a water channel, in rat brain. Treatments for 6h with 100-1000ng/ml HMGB1, not showing self-toxicity, of primary-cultured rat astrocytes didnot increase AQP4 mRNA, unexpectedly. In contrast, intracerebroventricular (i.c.v.) injection of 300ng of HMGB1 significantly increased AQP4 protein after 8h and formed edema after 24h in vivo. Thus, we investigated the roles of microglia as well as astrocytes. HMGB1 (1000ng/ml) drastically increased interleukin (IL)-1ß in the primary-cultured rat microglia after 2h. The exposure of microglia to conditioned medium with HMGB1 and 3mM adenosine 5'-triphosphate for 6h significantly increased AQP4 mRNA in astrocytes after 6h. Although 1000ng/ml HMGB1 didnot induce transfer of nuclear factor (NF)-κB into the nucleus in astrocytes after 1h, the conditioned medium containing IL-1ß led to its nuclear import. As factors likely to be involved in the nuclear import of NF-κB besides IL-1ß, nitric oxide and tumor necrosis factor-α didnot contribute under these conditions. Finally, i.c.v. injection of 30nmol parthenolide, an NF-κB inhibitor, reversed 300ng of HMGB1 injection-induced AQP4 protein increase after 8h in vivo. The effect of parthenolide and the outcomes obtained so far suggest that HMGB1 indirectly up-regulates AQP4 expression through diffusible factor(s) such as IL-1ß from microglia since HMGB1 by itself didnot affect NF-κB intracellular localization in astrocytes.


Assuntos
Aquaporina 4/fisiologia , Astrócitos/citologia , Proteínas HMGB/fisiologia , Microglia/citologia , Regulação para Cima/fisiologia , Animais , Aquaporina 4/genética , Astrócitos/metabolismo , Sequência de Bases , Primers do DNA , Interleucina-1beta/biossíntese , Microglia/metabolismo , NF-kappa B/metabolismo , RNA Mensageiro/genética , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real
13.
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
14.
Rev Iberoam Micol ; 31(1): 7-10, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24262630

RESUMO

Histoplasma capsulatum is a dimorphic fungal pathogen naturally found in the soil. Inhalation of conidia can result in pulmonary histoplasmosis and, in some cases, causes severe disseminated disease and death. This fungus is an ascomycete that has an anamorphic or asexual stage and a teleomorphic or sexual stage, known as Ajellomyces capsulatus, which results from (+) and (-) mating types. Sexual reproduction is regulated by a specialized genomic region known as the mating-type (MAT1) locus. The mating process in this heterothallic species is represented by isolates that contain only one of the two different MAT1 locus idiomorphs (MAT1-1 or MAT1-2) that have unrelated sequences encoding different transcription factors. In medically important dimorphic pathogens and in most ascomycete molds, one MAT locus idiomorph encodes a high-mobility-group (HMG) box-domain transcription factor, and the other idiomorph encodes an alpha-box domain transcription factor. There is scarce molecular information about H. capsulatum mating type although recombinant population structures have been reported that could occur in nature and this process has been documented in distinct models such as parasites and other fungi. In this review, we shall focus on published studies on H. capsulatum sexuality, and outline the distribution of the two H. capsulatum mating types in Latin America. This manuscript is part of the series of works presented at the "V International Workshop: Molecular genetic approaches to the study of human pathogenic fungi" (Oaxaca, Mexico, 2012).


Assuntos
Genes Fúngicos Tipo Acasalamento , Histoplasma/fisiologia , Brasil , Evolução Molecular , Proteínas Fúngicas/genética , Proteínas Fúngicas/fisiologia , Variação Genética , Proteínas HMGB/genética , Proteínas HMGB/fisiologia , Histoplasma/genética , Histoplasma/isolamento & purificação , Histoplasmose/microbiologia , Humanos , México , Reprodução , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia
15.
Fly (Austin) ; 6(4): 234-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22878648

RESUMO

Dorsoventral (DV) axis formation in Drosophila begins during oogenesis through the graded activation of the EGF receptor (EGFR)-Ras-MAPK signaling pathway in the follicle cell layer of the egg chamber. EGFR signaling, which is higher in dorsal follicle cells, represses expression of the sulfotransferase-encoding gene pipe, thereby delimiting a ventral domain of Pipe activity that is critical for the subsequent induction of ventral embryonic fates. We have characterized the transcriptional circuit that links EGFR signaling to pipe repression: in dorsal follicle cells, the homeodomain transcription factor Mirror (Mirr), which is induced by EGFR signaling, directly represses pipe transcription, whereas in ventral follicle cells, the HMG-box protein Capicua (Cic) supports pipe expression by repressing mirr. Although Cic is under negative post-transcriptional regulation by Ras-MAPK signaling in different contexts, the relevance of this mechanism for the interpretation of the EGFR signal during DV pattern formation remains unclear. Here, we consider a model where EGFR-mediated downregulation of Cic modulates the spatial distribution of Mirr protein in lateral follicle cells, thereby contributing to define the position at which the pipe expression border is formed.


Assuntos
Padronização Corporal/genética , Regulação para Baixo , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Drosophila/citologia , Receptores ErbB/fisiologia , Proteínas HMGB/genética , Receptores de Peptídeos de Invertebrados/fisiologia , Proteínas Repressoras/genética , Animais , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas HMGB/metabolismo , Proteínas HMGB/fisiologia , Modelos Biológicos , Proteínas Repressoras/metabolismo , Proteínas Repressoras/fisiologia , Transdução de Sinais , Sulfotransferases/genética , Sulfotransferases/metabolismo
16.
Mech Dev ; 129(9-12): 263-74, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22820002

RESUMO

The high mobility group (HMG) proteins constitute a superfamily of nuclear proteins that regulate the expression of a wide range of genes through architectural remodeling of the chromatin structure, and the formation of multiple protein complexes on promoter/enhancer regions, but their function in germ layer specification during early development is not clear. Here we show that hmgb genes regulate mesoderm formation and dorsoventral patterning both in zebrafish and Xenopus early embryos. Overexpression of hmgb3 blocks the expression of the pan-mesoderm gene no tail/Xbra and other ventrolateral mesoderm genes, and results in embryos with shortened anteroposterior axis, while overexpression of hmgb3EnR, which contains the engrailed repressor domain, most potently repressed no tail expression and mesoderm formation. However, hmgb3VP16, which contains the transcriptional activation domain of VP16, had an opposite effect, indicating that hmgb3 may function as a repressor during mesoderm induction and patterning. In addition, we show that hmgb3 inhibits target gene expression downstream of mesoderm-inducing factors. Furthermore, using reporter gene assays in Xenopus whole embryos, we show that hmgb3 differentially regulates the activation of various mesendoderm reporter genes. In particular, it up-regulates the goosecoid, but inhibits the Xbra reporter gene activation. Therefore, our results suggest that hmgb genes may function to fine-tune the specification and/or dorsoventral patterning of mesoderm during zebrafish and Xenopus development.


Assuntos
Padronização Corporal/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas HMGB/fisiologia , Xenopus/crescimento & desenvolvimento , Peixe-Zebra/crescimento & desenvolvimento , Animais , Padronização Corporal/genética , Proteínas Fetais , Proteína Goosecoid/genética , Proteína Goosecoid/metabolismo , Proteínas HMGB/genética , Proteínas HMGB/metabolismo , Proteína HMGB3/genética , Proteína HMGB3/metabolismo , Mesoderma/crescimento & desenvolvimento , Mesoderma/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Ativação Transcricional/genética , Regulação para Cima , Xenopus/genética , Xenopus/metabolismo , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
17.
Mol Endocrinol ; 26(6): 1014-27, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22543271

RESUMO

The pituitary is a complex gland comprising different cell types each secreting specific hormones. The extensive network of signaling molecules and transcription factors required for determination and terminal differentiation of specific cell types is still not fully understood. The SRY-like HMG-box (SOX) transcription factor Sox4 plays important roles in many developmental processes and has two homologs in zebrafish, Sox4a and Sox4b. We show that the sox4b gene is expressed in the pituitary anlagen starting at 24 h after fertilization (hpf) and later in the entire head region including the pituitary. At 48 hpf, sox4b mRNA colocalizes with that for TSH (tshß), glycoprotein subunit α (gsuα), and the Zn finger transcription factor Gata2a. Loss of Sox4b function, using morpholino knockdown or expression of a dominant-negative Sox4 mutant, leads to a drastic decrease in tshß and gsuα expression and reduced levels of gh, whereas other anterior pituitary gland markers including prl, slß, pomc, and lim3 are not affected. Sox4b is also required for expression of gata2a in the pituitary. Knockdown of gata2a leads to decreased tshß and gsuα expression at 48 hpf, similar to sox4b morphants. Injection of gata2a mRNA into sox4b morphants rescued tshß and gsuα expression in thyrotrope cells. Finally, sox4b or gata2a knockdown causes a significant decrease of gonadotropin expression (lhß and fshß) at 4 d after fertilization. In summary, our results indicate that Sox4b is expressed in zebrafish during pituitary development and plays a crucial role in the differentiation of thyrotrope and gonadotrope cells through induction of gata2a expression in the developing pituitary.


Assuntos
Fator de Transcrição GATA2/metabolismo , Gonadotrofos/fisiologia , Proteínas HMGB/fisiologia , Adeno-Hipófise/metabolismo , Tireotrofos/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/metabolismo , Análise de Variância , Animais , Subunidade beta do Hormônio Folículoestimulante/genética , Subunidade beta do Hormônio Folículoestimulante/metabolismo , Fator de Transcrição GATA2/genética , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Subunidade alfa de Hormônios Glicoproteicos/genética , Subunidade alfa de Hormônios Glicoproteicos/metabolismo , Gonadotrofos/metabolismo , Proteínas HMGB/genética , Proteínas HMGB/metabolismo , Hormônio Luteinizante Subunidade beta/genética , Hormônio Luteinizante Subunidade beta/metabolismo , Morfolinos/genética , Mucolipidoses , Organogênese , Adeno-Hipófise/citologia , Adeno-Hipófise/embriologia , Tireotrofos/metabolismo , Tireotropina Subunidade beta/genética , Tireotropina Subunidade beta/metabolismo , Transcrição Gênica , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
18.
J Neurosci ; 31(39): 13921-35, 2011 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-21957254

RESUMO

The SRY-box (Sox) transcription factors regulate oligodendrocyte differentiation, but their signaling targets are largely unknown. We have identified a major signal transduction pathway regulated by Sox containing gene 17 (Sox17) in the oligodendrocyte lineage. Microarray analysis in oligodendrocyte progenitor cells (OPCs) after Sox17 attenuation revealed upregulated genes associated with cell cycle control and activation of the Wingless and integration site (Wnt)/ß-catenin pathway. Sox17 knockdown also increases the levels of cyclin D1, Axin2, and activated ß-catenin. In OPCs, the expression pattern of Sox17, cyclin D1, and secreted Frizzled-related protein-1 in the presence of platelet-derived growth factor (PDGF) was coordinately accelerated by addition of thyroid hormone, indicating differentiation-induced regulation of Sox17 targets. In developing white matter, decreased total ß-catenin, activated ß-catenin, and cyclin D1 levels coincided with the peak of Sox17 expression, and immunoprecipitates showed a developmentally regulated interaction among Sox17, T-cell transcription factor 4, and ß-catenin proteins. In OPCs, PDGF stimulated phosphorylation of glycogen synthase 3ß and the Wnt coreceptor LRP6, and enhanced ß-catenin-dependent gene expression. Sox17 overexpression inhibited PDGF-induced TOPFLASH and cyclin D1 promoter activity, and decreased endogenous cyclin D1, activated ß-catenin, as well as total ß-catenin levels. Recombinant Sox17 prevented Wnt3a from repressing myelin protein expression, and inhibition of Sox17-mediated proteasomal degradation of ß-catenin blocked myelin protein induction. These results indicate that Sox17 suppresses cyclin D1 expression and cell proliferation by directly antagonizing ß-catenin, whose activity in OPCs is stimulated not only by Wnt3a, but also by PDGF. Our identification of downstream targets of Sox17 thus defines signaling pathways and molecular mechanisms in OPCs that are regulated by Sox17 during cell cycle exit and the onset of differentiation in oligodendrocyte development.


Assuntos
Proteínas HMGB/fisiologia , Oligodendroglia/fisiologia , Fatores de Transcrição SOXF/fisiologia , Transdução de Sinais/fisiologia , Células-Tronco/fisiologia , Proteínas Wnt/fisiologia , beta Catenina/fisiologia , Animais , Células Cultivadas , Técnicas de Introdução de Genes , Proteínas HMGB/antagonistas & inibidores , Proteínas HMGB/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas da Mielina/antagonistas & inibidores , Proteínas da Mielina/biossíntese , Células NIH 3T3 , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Fatores de Transcrição SOXF/antagonistas & inibidores , Fatores de Transcrição SOXF/genética , beta Catenina/antagonistas & inibidores
19.
J Intern Med ; 270(4): 301-8, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21793952

RESUMO

Central to protective and pathological immunity is the activation of innate immune responses upon recognition of nucleic acids by transmembrane Toll-like receptors (TLRs) and cytosolic receptors. In mammals, the transmembrane pattern recognition receptors TLR3, TLR7 and TLR9 recognize double-stranded RNA, single-stranded RNA and hypomethylated DNA, respectively, while the retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs), RIG-I and MDA5 are known to be cytosolic RNA-sensing receptors. In addition, cytosolic DNA-sensing receptors that include DAI, RIG-I/MDA5 and AIM2 also trigger innate immune responses. High-mobility group box (HMGB)1, 2 and 3 proteins, which also bind immunogenic nucleic acids, are generally involved in the nucleic acid receptor-mediated activation of innate immune responses. There is a hierarchy in the nucleic acid-mediated activation of immune responses, wherein the selective activation of the nucleic acid-sensing receptors is contingent on the more promiscuous sensing of nucleic acids by HMGBs. The aim of this review is to summarize this novel feature of HMGB proteins, as essential frontline instigators of nucleic acid-mediated activation of innate immune responses. In addition, we will discuss the therapeutic implications of these findings.


Assuntos
Proteínas HMGB/fisiologia , Imunidade Inata/imunologia , Transdução de Sinais/imunologia , Receptores Toll-Like/imunologia , DNA/imunologia , Humanos , Ácidos Nucleicos/imunologia , RNA/imunologia
20.
Cancer Sci ; 102(7): 1313-21, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21457403

RESUMO

CD133 is a universal marker of tissue stem/progenitor cells as well as cancer stem cells, but its physiological significance remains to be elucidated. Here we examined the relationship between expression of CD133 and features of gastric epithelial cells, and found that CD133-positive (CD133[+]) tumor cell lines formed well-differentiated tumors while CD133-negative (CD133[-]) lines formed poorly differentiated ones when subcutaneously injected into nude mice. We also found that CD133(+) and CD133(-) cell populations co-existed in some cell lines. FACS analysis showed that CD133(+) cells were mother cells because CD133(+) cells formed both CD133(+) and CD133(-) cells, but CD133(-) cells did not form CD133(+) cells. In these cell lines, CD133(+) cells formed well-differentiated tumors while CD133(-) cells formed poorly differentiated ones. In human gastric cancers, CD133 was exclusively expressed on the luminal surface membrane of gland-forming cells, and it was never found on poorly differentiated diffuse-type cells. Considering that poorly differentiated tumors often develop from well-differentiated tumors during tumor progression, these results suggest that loss of expression of CD133 might be related to gastric tumor progression. Microarray analysis showed that CD133(+) cells specifically expressed Sox17, a tumor suppressor in gastric carcinogenesis. Forced expression of SOX17 induced expression of CD133 in CD133(-) cells, and reduction of SOX17 caused by siRNA in CD133(+) cells induced a reduction in the level of CD133. These results indicate that Sox17 might be a key transcription factor controlling CD133 expression, and that it might also play a role in the control of gastric tumor progression.


Assuntos
Antígenos CD/fisiologia , Glicoproteínas/fisiologia , Proteínas HMGB/fisiologia , Células-Tronco Neoplásicas/química , Peptídeos/fisiologia , Fatores de Transcrição SOXF/fisiologia , Neoplasias Gástricas/patologia , Antígeno AC133 , Animais , Antígenos CD/análise , Feminino , Mucosa Gástrica/patologia , Perfilação da Expressão Gênica , Glicoproteínas/análise , Proteínas HMGB/análise , Humanos , Camundongos , Peptídeos/análise , Fatores de Transcrição SOXF/análise , Neoplasias Gástricas/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA