Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Gan To Kagaku Ryoho ; 51(3): 231-236, 2024 Mar.
Artigo em Japonês | MEDLINE | ID: mdl-38494798

RESUMO

Return of individual genomic results(ROGR)to participants in population-based biobank has been rarely conducted in research settings, and the procedure of ROGR performed in foreign countries may not be simply applied to Japanese participants, because of the difference in social background. The Tohoku Medical Megabank Project, which was launched in 2012 aiming to build a foundation of personalized genomic medicine, obtained the consent from research participants by explaining the future possibility of ROGR. After careful consideration of appropriate procedure for ROGR, individual genomic results were returned to 111 pathogenic variant(PV)carriers of hereditary breast and ovarian cancer syndrome(HBOC)or Lynch syndrome(LS)based on 50,000 whole genome sequencing(WGS)data in FY 2022. Since majority of the participants has no cancer diagnosis, participants' right to not know was carefully considered. In addition, the variants to be returned were carefully evaluated by using multiple databases, and the WGS results of the participants were further confirmed by the single- site analysis. When the genomic results were returned, the participants were informed about clinical risk surveillance at the hospital. Seventy-eight and 33 PV carriers of HBOC and LS, respectively, participated in the study. Most participants were in their 30s or 40s. Unexpectedly, validation testing results of 12 LS participants were found to be negative or variant of uncertain significance, because detecting these variants by WGS were technically challenging. Twenty-eight participants (HBOC 20, LS 8)had been diagnosed as cancer, and 6 females who had breast cancer were genetically diagnosed as HBOC and underwent or planned risk-reducing surgery. Eighteen participants refused to undergo clinical risk surveillance because of the medical expense that is not covered by health insurance and the burden of visiting the hospital. The opportunity to undergo medical surveillance should be provided to population-based cohort participants who carry actionable pathogenic variants, and ROGR to general population should be promoted to create the base of personalized genomic medicine.


Assuntos
Síndrome Hereditária de Câncer de Mama e Ovário , Feminino , Humanos , Estudos de Coortes , Predisposição Genética para Doença , Genômica , Células Germinativas
2.
Breast Cancer ; 30(1): 110-120, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36161580

RESUMO

BACKGROUND: Recent advances in human genome research have provided evidence for genotype-phenotype associations, pathogenicity, and clinical actionability of variants and genomic risk prediction of disease. However, the return of individual genomic results to healthy individuals is fraught with ethical and practical complexity. METHODS: Individual genomic results were returned to BRCA1/2 pathogenic variant (PV) carriers of the Tohoku Medical Megabank cohort study participants with an information on hereditary breast and ovarian cancer syndrome (HBOC). One hundred and eighty participants, including 9 BRCA1/2 PV carriers, were asked about their willingness to receive individual genomic results, without revealing the gene name and related disorders, prior to the study. Of the 142 participants who responded, 103 showed willingness to know their genomic information. Each of the six BRCA1/2 PV carriers who consented to participate in the study received information about HBOC in person and underwent validation testing with blood resampling. RESULTS: All participants were in their 60s or 70s; of the four females and two males, two had a history of breast cancer and five had a family history of HBOC-related cancers. All participants appreciated the information, without remarkable negative psychological impact of the return, and intended to undergo clinical risk surveillance. Five participants were accompanied by family members while receiving the results, and three first-degree female relatives wished to undergo genomic testing at the hospital. CONCLUSIONS: Our results suggest that returning actionable genomic information to participants in a population-based genome cohort study is beneficial for preventing or providing early-stage intervention for associated diseases.


Assuntos
Neoplasias da Mama , Síndrome Hereditária de Câncer de Mama e Ovário , Neoplasias Ovarianas , Masculino , Humanos , Feminino , Neoplasias da Mama/genética , Estudos de Coortes , Proteína BRCA1/genética , Síndrome Hereditária de Câncer de Mama e Ovário/genética , Genômica , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/prevenção & controle , Predisposição Genética para Doença , Proteína BRCA2/genética
3.
J Hum Genet ; 67(1): 9-17, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34234266

RESUMO

Certain large genome cohort studies attempt to return the individual genomic results to the participants; however, the implementation process and psychosocial impacts remain largely unknown. The Tohoku Medical Megabank Project has conducted large genome cohort studies of general residents. To implement the disclosure of individual genomic results, we extracted the potential challenges and obstacles. Major challenges include the determination of genes/disorders based on the current medical system in Japan, the storage of results, prevention of misunderstanding, and collaboration of medical professionals. To overcome these challenges, we plan to conduct multilayer pilot studies, which deal with different disorders/genes. We finally chose familial hypercholesterolemia (FH) as a target disease for the first pilot study. Of the 665 eligible candidates, 33.5% were interested in the pilot study and provided consent after an educational "genetics workshop" on the basic genetics and medical facts of FH. The genetics professionals disclosed the results to the participants. All positive participants were referred to medical care, and a serial questionnaire revealed no significant psychosocial distress after the disclosure. Return of genomic results to research participants was implemented using a well-prepared protocol. To further elucidate the impact of different disorders, we will perform multilayer pilot studies with different disorders, including actionable pharmacogenomics and hereditary tumor syndromes.


Assuntos
Genética Médica , Genoma , Genômica , Pesquisa , Bases de Dados Genéticas , Revelação , Genômica/métodos , Humanos , Japão , Farmacogenética , Projetos Piloto , Projetos de Pesquisa
4.
Sci Rep ; 10(1): 17315, 2020 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-33057147

RESUMO

Aging induces numerous cellular disorders, such as the elevation of reactive oxygen species (ROS), in a number type of cells, including mesenchymal stem cells (MSCs). However, the correlation of ROS and impaired healing abilities as well as whether or not the inhibition of elevating ROS results in the rejuvenation of elderly MSCs is unclear. The rejuvenation of aged MSCs has thus recently received attention in the field of regenerative medicine. Specifically, extracellular vesicles (EVs) act as a novel tool for stem cell rejuvenation due to their gene transfer ability with systemic effects and safety. In the present study, we examined the roles of aging-associated ROS in the function and rejuvenation of elderly MSCs by infant EVs. The data clearly showed that elderly MSCs exhibited the downregulation of superoxide dismutase (SOD)1 and SOD3, which resulted in the elevation of ROS and downregulation of the MEK/ERK pathways, which are involved in the impairment of the MSCs' ability to decrease necrotic area in the skin flap model. Furthermore, treatment with the antioxidant Edaravone or co-overexpression of SOD1 and SOD3 rescued elderly MSCs from the elevation of ROS and cellular senescence, thereby improving their functions. Of note, infant MSC-derived EVs rejuvenated elderly MSCs by inhibiting ROS production and the acceleration of cellular senescence and promoting the proliferation and in vivo functions in both type 1 and type 2 diabetic mice.


Assuntos
Vesículas Extracelulares/fisiologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Espécies Reativas de Oxigênio/metabolismo , Rejuvenescimento/fisiologia , Envelhecimento/metabolismo , Animais , Senescência Celular/fisiologia , Diabetes Mellitus Experimental/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases , Células-Tronco Mesenquimais/metabolismo , Camundongos , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1/metabolismo
5.
Int J Mol Sci ; 20(18)2019 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-31533351

RESUMO

Mast cell tryptases have crucial roles in allergic and inflammatory diseases. The mouse tryptase genes represent a cluster of loci on chromosome 16p3.3. While their functional studies have been extensively performed, transcriptional regulation of tryptase genes is poorly understood. In this study, we examined the molecular basis of the tryptase gene expression in bone marrow-derived mast cells (BMMCs) of C57BL/6 mice and in MEDMC-BRC6 mast cells. The expression of the Tpsb2 and Tpsg1 genes, which reside at the 3'-end of the tryptase locus, is significantly decreased by the reduction of the GATA transcription factors GATA1 or GATA2. Chromatin immunoprecipitation assays have shown that the GATA factors bind at multiple regions within the locus, including 1.0 and 72.8 kb upstream of the Tpsb2 gene, and that GATA1 and GATA2 facilitate each other's DNA binding activity to these regions. Deletion of the -72.8 kb region by genome editing significantly reduced the Tpsb2 and Tpsg1 mRNA levels in MEDMC-BRC6 cells. Furthermore, binding of CTCF and the cohesin subunit Rad21 was found upstream of the -72.8 kb region and was significantly reduced in the absence of GATA1. These results suggest that mouse tryptase gene expression is coordinately regulated by GATA1 and GATA2 in BMMCs.


Assuntos
Células da Medula Óssea/metabolismo , Fator de Transcrição GATA1/metabolismo , Fator de Transcrição GATA2/metabolismo , Regulação da Expressão Gênica , Mastócitos/metabolismo , Triptases/genética , Animais , Técnicas de Inativação de Genes , Loci Gênicos , Camundongos , Regiões Promotoras Genéticas , Ligação Proteica , RNA Interferente Pequeno/genética , Triptases/metabolismo
6.
Mol Cell Biol ; 39(22)2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31501274

RESUMO

GATA factors GATA1 and GATA2 and ETS factor PU.1 are known to function antagonistically during hematopoietic development. In mouse mast cells, however, these factors are coexpressed and activate the expression of the Ms4a2 gene encoding the ß chain of the high-affinity IgE receptor (FcεRI). The present study showed that these factors cooperatively regulate Ms4a2 gene expression through distinct mechanisms. Although GATA2 and PU.1 contributed almost equally to Ms4a2 gene expression, gene ablation experiments revealed that simultaneous knockdown of both factors showed neither a synergistic nor an additive effect. A chromatin immunoprecipitation analysis showed that they shared DNA binding to the +10.4-kbp region downstream of the Ms4a2 gene with chromatin looping factor LDB1, whereas the proximal -60-bp region was exclusively bound by GATA2 in a mast cell-specific manner. Ablation of PU.1 significantly reduced the level of GATA2 binding to both the +10.4-kbp and -60-bp regions. Surprisingly, the deletion of the +10.4-kbp region by genome editing completely abolished the Ms4a2 gene expression as well as the cell surface expression of FcεRI. These results suggest that PU.1 and LDB1 play central roles in the formation of active chromatin structure whereas GATA2 directly activates the Ms4a2 promoter.


Assuntos
Fator de Transcrição GATA2/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores de IgE/genética , Transativadores/metabolismo , Animais , Células da Medula Óssea/citologia , Imunoprecipitação da Cromatina , Proteínas de Ligação a DNA/metabolismo , Fator de Transcrição GATA2/genética , Regulação da Expressão Gênica , Proteínas com Domínio LIM/metabolismo , Mastócitos/metabolismo , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/genética , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Receptores de IgE/metabolismo , Transativadores/genética
7.
Stem Cells Dev ; 26(13): 948-963, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28537846

RESUMO

Chronic kidney disease (CKD) results in a delay in wound healing because of its complications such as uremia, anemia, and fluid overload. Mesenchymal stem cells (MSCs) are considered to be a candidate for wound healing because of the ability to recruit many types of cells. However, it is still unclear whether the CKD-adipose tissue-derived MSCs (CKD-AT-MSCs) have the same function in wound healing as healthy donor-derived normal AT-MSCs (nAT-MSCs). In this study, we found that uremic toxins induced elevated reactive oxygen species (ROS) expression in nAT-MSCs, resulting in the reduced expression of hypoxia-inducible factor-1α (HIF-1α) under hypoxic conditions. Consistent with the uremic-treated AT-MSCs, there was a definite imbalance of redox state and high expression of ROS in CKD-AT-MSCs isolated from early-stage CKD patients. In addition, a transplantation study clearly revealed that nAT-MSCs promoted the recruitment of inflammatory cells and recovery from ischemia in the mouse flap model, whereas CKD-AT-MSCs had defective functions and the wound healing process was delayed. Of note, the expression of prolyl hydroxylase domain 2 (PHD2) is selectively increased in CKD-AT-MSCs and its inhibition can restore the expression of HIF-1α and the wound healing function of CKD-AT-MSCs. These results indicate that more studies about the functions of MSCs from CKD patients are required before they can be applied in the clinical setting.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Células-Tronco Mesenquimais/metabolismo , Cicatrização/genética , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Animais , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia/genética , Transplante de Células-Tronco Mesenquimais , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Insuficiência Renal Crônica/metabolismo , Cicatrização/fisiologia
8.
Biochem Biophys Res Commun ; 477(1): 68-75, 2016 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-27282479

RESUMO

Mesenchymal stem cells (MSCs) are defined as multipotent cells that can give rise to various kinds of differentiated mesenchymal cells, and are thus considered to be useful for clinical therapy. However, the big hurdles of MSC therapy are the inability of MSCs to reach the appropriate tissues or sites with high efficiency and engraftment after transplantation. In this study, we investigated how adipose tissue-derived MSCs (AT-MSCs) improve their homing ability after intravenous injection. We previously found that human endothelial progenitor cells with low aldehyde dehydrogenase activity (Alde-Low EPCs) are suitable for the treatment of ischemic tissues. In addition, we demonstrated that microvesicles (MVs) derived from Alde-Low EPCs possessed the ability to improve the homing ability of non-functional Alde-High EPCs, resulting in wound healing. We initially transfected MVs derived from Alde-Low EPCs (EMVs) to human AT-MSCs, which were originally unable to cure ischemic tissues by intravenous transplantation. Remarkably, AT-MSC transfected EMVs dramatically repaired the ischemic skin flap compared with AT-MSC derived-MV (MMVs) transfected AT-MSCs or control AT-MSCs. We then found that the expression of CXCR4, an important chemokine receptor for cell migration, was highly elevated in EMV-transfected AT-MSCs. Moreover, AT-MSCs transfected with EMVs, but not control AT-MSCs, migrated to wound sites after intravenous injection. Consequently, CD45(+) inflammatory cells were successfully recruited at the wound sites after the injection of EMV-transfected AT-MSCs. These results demonstrate that EMVs are a useful source to improve the homing ability and wound healing ability of MSCs at the wound sites.


Assuntos
Células Progenitoras Endoteliais/citologia , Células-Tronco Mesenquimais/citologia , Cicatrização , Animais , Camundongos , Camundongos Endogâmicos C57BL
9.
Biochem Biophys Res Commun ; 473(4): 1111-1118, 2016 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-27063802

RESUMO

Microvesicles (MVs) derived from mesenchymal stem cells showed the ability to alter the cell phenotype and function. We previously demonstrated that type 2 diabetic adipose tissue-derived mesenchymal stem cells (dAT-MSCs) increase in cell aggregation and adhesion in vitro and impair wound healing in vivo. However, the characterization and function of MVs derived from human non-diabetic AT-MSCs (nAT-MSCs) remain unknown. In this study, we characterized nAT-MSC-derived MVs and their function after the transfection of dAT-MSCs with MVs using the scratch assay and a flap mouse model. We found that human nAT-MSC-derived MVs expressed MSC-surface markers and improved dAT-MSC functions by altering the expression of genes associated with cell migration, survival, inflammation, and angiogenesis as well as miR29c and miR150. Remarkably, the transfection of dAT-MSCs with nAT-MSC-derived MVs improved their migration ability in vitro and wound healing ability in a flap mouse model. These results demonstrate a promising opportunity to modify the function of dAT-MSCs for therapeutic stem cell application in diabetic patients.


Assuntos
Adipócitos/citologia , Micropartículas Derivadas de Células/transplante , Diabetes Mellitus Tipo 2/patologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/fisiologia , Cicatrização/fisiologia , Animais , Movimento Celular , Micropartículas Derivadas de Células/patologia , Micropartículas Derivadas de Células/fisiologia , Feminino , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Resultado do Tratamento
10.
Stem Cells Dev ; 25(10): 760-73, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-26988763

RESUMO

The prevalence of type 2 diabetes mellitus (T2DM), which leads to diabetic complications, has been increasing worldwide. The possible applications of T2DM-derived stem cells in cell therapy are limited because their characteristics are still not fully understood. In this study, we characterized adipose tissue-derived mesenchymal stem cells (AT-MSCs) from diabetic patients (dAT-MSCs) and found that insulin receptor substrate-1 (IRS-1) was highly phosphorylated at serine 636/639 in dAT-MSCs. Moreover, we found that early growth response factor-1 (EGR-1) and its target genes of PTEN and GGPS1 were highly expressed in dAT-MSCs in comparison to healthy donor-derived AT-MSCs (nAT-MSCs). We observed impaired wound healing after the injection of dAT-MSCs in the ischemic flap mouse model. The expressions of EGR-1 and its target genes were diminished by small hairpin RNA-targeted EGR-1 (shEGR-1) and treatment with a mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) inhibitor (PD98059). Importantly, dAT-MSCs with shEGR-1 were able to restore the wound healing ability in the mouse model. Interestingly, under hypoxic conditions, hypoxia-inducible factor-1α (HIF-1α) can bind to the EGR-1 promoter in dAT-MSCs, but not in nAT-MSCs. Together, these results demonstrate that the expression of EGR-1 was upregulated in dAT-MSCs through two pathways: the main regulatory pathway is the MAPK/ERK pathway, the other is mediated by HIF-1α through direct transcriptional activation at the promoter region of the EGR1 gene. Our study suggests that dAT-MSCs may contribute to microvascular damage and delay wound healing through the overexpression of EGR-1. Interrupting the expression of EGR-1 in dAT-MSCs may be a useful treatment for chronic wounds in diabetic patients.


Assuntos
Tecido Adiposo/patologia , Diabetes Mellitus Tipo 2/patologia , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Células-Tronco Mesenquimais/metabolismo , Cicatrização , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Membrana Celular/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Pele/patologia , Retalhos Cirúrgicos
11.
Blood ; 125(21): 3306-15, 2015 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-25855601

RESUMO

GATA2 plays a crucial role for the mast cell fate decision. We herein demonstrate that GATA2 is also required for the maintenance of the cellular identity in committed mast cells derived from mouse bone marrow (BMMCs). The deletion of the GATA2 DNA binding domain (GATA2ΔCF) in BMMCs resulted in a loss of the mast cell phenotype and an increase in the number of CD11b- and/or Ly6G/C-positive cells. These cells showed the ability to differentiate into macrophage- and neutrophil-like cells but not into eosinophils. Although the mRNA levels of basophil-specific genes were elevated, CD49b, a representative basophil marker, never appeared on these cells. GATA2 ablation led to a significant upregulation of C/EBPα, and forced expression of C/EBPα in wild-type BMMCs phenocopied the GATA2ΔCF cells. Interestingly, simultaneous deletion of the Gata2 and Cebpa genes in BMMCs restored the aberrant increases of CD11b and Ly6G/C while retaining the reduced c-Kit expression. Chromatin immunoprecipitation assays indicated that GATA2 directly binds to the +37-kb region of the Cebpa gene and thereby inhibits the RUNX1 and PU.1 binding to the neighboring region. Upregulation of C/EBPα following the loss of GATA2 was not observed in cultured mast cells derived from peritoneal fluid, whereas the repression of c-Kit and other mast cell-specific genes were observed in these cells. Collectively, these results indicate that GATA2 maintains cellular identity by preventing Cebpa gene activation in a subpopulation of mast cells, whereas it plays a fundamental role as a positive regulator of mast cell-specific genes throughout development of this cell lineage.


Assuntos
Células da Medula Óssea/citologia , Desdiferenciação Celular/imunologia , Fator de Transcrição GATA2/metabolismo , Mastócitos/citologia , Células-Tronco/citologia , Animais , Western Blotting , Diferenciação Celular/imunologia , Imunoprecipitação da Cromatina , Citometria de Fluxo , Fator de Transcrição GATA2/imunologia , Mastócitos/metabolismo , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Mol Cell Biol ; 35(10): 1825-37, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25755285

RESUMO

GATA1 is a key transcription factor for erythropoiesis. GATA1 gene expression is strictly regulated at the transcriptional level. While the regulatory mechanisms governing mouse Gata1 (mGata1) gene expression have been studied extensively, how expression of the human GATA1 (hGATA1) gene is regulated remains to be elucidated. To address this issue, we generated hGATA1 bacterial artificial chromosome (BAC) transgenic mouse lines harboring a 183-kb hGATA1 locus covering the hGATA1 exons and distal flanking sequences. Transgenic hGATA1 expression coincides with endogenous mGata1 expression and fully rescues hematopoietic deficiency in mGata1 knockdown mice. The transgene exhibited copy number-dependent and integration position-independent expression of hGATA1, indicating the presence of chromatin insulator activity within the transgene. We found a novel insulator element at 29 kb 5' to the hGATA1 gene and refer to this element as the 5' CCCTC-binding factor (CTCF) site. Substitution mutation of the 5' CTCF site in the hGATA1 BAC disrupted the chromatin architecture and led to a reduction of hGATA1 expression in splenic erythroblasts under conditions of stress erythropoiesis. Our results demonstrate that expression of the hGATA1 gene is regulated through the chromatin architecture organized by 5' CTCF site-mediated intrachromosomal interactions in the hGATA1 locus.


Assuntos
Cromossomos/genética , Eritroblastos/metabolismo , Fator de Transcrição GATA1/genética , Elementos Isolantes , Baço/citologia , Animais , Sítios de Ligação , Fator de Ligação a CCCTC , Células Cultivadas , Cromatina/fisiologia , Cromossomos Artificiais Bacterianos/genética , Fator de Transcrição GATA1/química , Fator de Transcrição GATA1/metabolismo , Vetores Genéticos/genética , Humanos , Células K562 , Camundongos , Camundongos Transgênicos , Proteínas Repressoras/metabolismo , Baço/metabolismo
13.
Genes Cells ; 20(3): 224-41, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25626335

RESUMO

The variants of the hypoxia-inducible factor-3α gene HIF-3α and NEPAS are known to repress the transcriptional activities driven by HIF-1α and HIF-2α. Although NEPAS has been shown to play an important role in vascular remodeling during lung development, little is known about the roles of HIF-3α in adult lung function. Here, we examined pulmonary endothelial cells (ECs) isolated from wild-type (WT) and HIF-3α functional knockout (KO) mice. The expression levels of angiogenic factors (Flk1, Ang2 and Tie2) were significantly greater in the HIF-3α KO ECs than those in the WT ECs irrespective of oxygen tension. However, the HIF-3α KO ECs showed impaired proliferative and angiogenic activities. The impaired EC function was likely due to the excess vascular endothelial (VE)-cadherin, an inhibitor of Flk1/PI3 kinase/Akt signaling, as treatment of the cells to a neutralizing antibody partly restored the phenotype of the HIF-3α KO ECs. Importantly, we found that the mRNA levels of HIF-2α and Ets-1 were significantly increased by HIF-3α ablation. Given that both factors are known to activate the VE-cadherin gene, the transcriptional repression of these factors by HIF-3α might be important for silencing the irrelevant expression of the VE-cadherin gene. Collectively, these data show novel and unique roles of HIF-3α for angiogenic gene regulation in pulmonary ECs.


Assuntos
Antígenos CD/metabolismo , Caderinas/metabolismo , Células Endoteliais/metabolismo , Pulmão/citologia , Neovascularização Fisiológica , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Angiogênicas/metabolismo , Animais , Antígenos CD/genética , Proteínas Reguladoras de Apoptose , Caderinas/genética , Hipóxia Celular , Pulmão/irrigação sanguínea , Camundongos , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Repressoras , Fator A de Crescimento do Endotélio Vascular/metabolismo
14.
Mol Cell Biol ; 35(5): 805-15, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25535330

RESUMO

GATA1 is a master regulator of erythropoiesis, expression of which is regulated by multiple discrete cis-acting elements. In this study, we examine the activity of a promoter-proximal double GATA (dbGATA) motif, using a Gata1 bacterial artificial chromosome (BAC)-transgenic green fluorescent protein (GFP) reporter (G1BAC-GFP) mouse system. Deletion of the dbGATA motif led to significant reductions in GFP expression in hematopoietic progenitors, while GFP expression was maintained in erythroblasts. Consistently, in mice with a germ line deletion of the dbGATA motif (Gata1(ΔdbGATA) mice), GATA1 expression in progenitors was significantly decreased. The suppressed GATA1 expression was associated with a compensatory increase in GATA2 levels in progenitors. When we crossed Gata1(ΔdbGATA) mice with Gata2 hypomorphic mutant mice (Gata2(fGN/fGN) mice), the Gata1(ΔdbGATA)::Gata2(fGN/fGN) compound mutant mice succumbed to a significant decrease in the progenitor population, whereas both groups of single mutant mice maintained progenitors and survived to adulthood, indicating the functional redundancy between GATA1 and GATA2 in progenitors. Meanwhile, the effects of the dbGATA site deletion on Gata1 expression were subtle in erythroblasts, which showed increased GATA1 binding and enhanced accumulation of active histone marks around the 1st-intron GATA motif of the ΔdbGATA locus. These results thus reveal a novel role of the dbGATA motif in the maintenance of Gata1 expression in hematopoietic progenitors and a functional compensation between the dbGATA site and the 1st-intron GATA motif in erythroblasts.


Assuntos
Eritroblastos/citologia , Fator de Transcrição GATA1/metabolismo , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Alelos , Motivos de Aminoácidos , Animais , Separação Celular , Imunoprecipitação da Cromatina , Cromossomos Artificiais Bacterianos , Eritropoese , Feminino , Citometria de Fluxo , Fator de Transcrição GATA2/metabolismo , Deleção de Genes , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Homeostase , Íntrons , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Ligação Proteica
15.
Mol Cell Biol ; 34(10): 1812-26, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24615013

RESUMO

Although previous studies have shown that GATA1 is required for mast cell differentiation, the effects of the complete ablation of GATA1 in mast cells have not been examined. Using conditional Gata1 knockout mice (Gata1(-/y)), we demonstrate here that the complete ablation of GATA1 has a minimal effect on the number and distribution of peripheral tissue mast cells in adult mice. The Gata1(-/y) bone marrow cells were capable of differentiating into mast cells ex vivo. Microarray analyses showed that the repression of GATA1 in bone marrow mast cells (BMMCs) has a small impact on the mast cell-specific gene expression in most cases. Interestingly, however, the expression levels of mast cell tryptases in the mouse chromosome 17A3.3 were uniformly reduced in the GATA1 knockdown cells, and GATA1 was found to bind to a 500-bp region at the 5' end of this locus. Revealing a sharp contrast to that observed in the Gata1-null BMMCs, GATA2 deficiency resulted in a significant loss of the c-Kit(+) FcεRIα(+) mast cell fraction and a reduced expression of several mast cell-specific genes. Collectively, GATA2 plays a more important role than GATA1 in the regulation of most mast cell-specific genes, while GATA1 might play specific roles in mast cell functions.


Assuntos
Diferenciação Celular , Fator de Transcrição GATA1/fisiologia , Fator de Transcrição GATA2/fisiologia , Mastócitos/fisiologia , Animais , Sequência de Bases , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Células Cultivadas , Cromossomos de Mamíferos , Meios de Cultura , Regulação Enzimológica da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Transcriptoma , Triptases/genética , Triptases/metabolismo
16.
Blood ; 122(20): 3450-60, 2013 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-24021675

RESUMO

GATA1 is a master regulator of hematopoietic differentiation, but Gata1 expression is inactivated in hematopoietic stem cells (HSCs). Using a bacterial artificial chromosome containing the Gata1 gene modified with green fluorescent protein (GFP) reporter, we explored the function of the 3.7-kb Gata1 upstream region (GdC region) that harbors 3 core cis-elements: Gata1 hematopoietic enhancer, double GATA-motif, and CACCC-motif. Transgenic GFP expression directed by the Gata1-BAC faithfully recapitulated the endogenous Gata1 expression pattern. However, deletion of the GdC-region eliminated reporter expression in all hematopoietic cells. To test whether the combination of the core cis-elements represents the regulatory function of the GdC-region, we replaced the region with a 659-bp minigene that linked the three cis-elements (MG-GFP). The GFP reporter expression directed by the MG-GFP BAC fully recapitulated the erythroid-megakaryocytic Gata1 expression. However, the GFP expression was aberrantly increased in the HSCs and was associated with decreases in DNA methylation and abundant GATA2 binding to the transgenic MG-GFP allele. The 3.2-kb sequences interspaced between the Gata1 hematopoietic enhancer and the double GATA-motif were able to recruit DNA methyltransferase 1, thereby exerting a cis-repressive function in the HSC-like cell line. These results indicate that the 3.2-kb interspacing sequences inactivate Gata1 by maintaining DNA-methylation in the HSCs.


Assuntos
Células Eritroides/metabolismo , Fator de Transcrição GATA1/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Células-Tronco Hematopoéticas/metabolismo , Sequências Reguladoras de Ácido Nucleico/genética , Animais , Linhagem da Célula , Células Cultivadas/metabolismo , Cromossomos Artificiais Bacterianos , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA/genética , Elementos Facilitadores Genéticos/genética , Eritropoese/genética , Fator de Transcrição GATA1/fisiologia , Fator de Transcrição GATA2/metabolismo , Inativação Gênica , Genes Reporter , Genes Sintéticos , Fígado/citologia , Fígado/embriologia , Megacariócitos/metabolismo , Camundongos , Camundongos Transgênicos , Motivos de Nucleotídeos/genética , Deleção de Sequência , Ativação Transcricional/genética
17.
Genes Cells ; 18(11): 921-33, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23911012

RESUMO

Transcription factor GATA2 is highly expressed in hematopoietic stem cells and progenitors, whereas its expression declines after erythroid commitment of progenitors. In contrast, the start of GATA1 expression coincides with the erythroid commitment and increases along with the erythroid differentiation. We refer this dynamic transition of GATA factor expression to as the 'GATA factor switching'. Here, we examined contribution of the GATA factor switching to the erythroid differentiation. In Gata1-knockdown embryos that concomitantly express Gata2-GFP reporter, high-level expression of GFP reporter was detected in accumulated immature hematopoietic cells with impaired differentiation, demonstrating that GATA1 represses Gata2 gene expression in hematopoietic progenitors in vivo. We have conducted chromatin immunoprecipitation (ChIP) on microarray analyses of GATA2 and GATA1, and results indicate that the GATA1-binding sites widely overlap with the sites pre-occupied by GATA2 before the GATA1 expression. Importantly, erythroid genes harboring GATA boxes bound by both GATA1 and GATA2 tend to be expressed in immature erythroid cells, whereas those harboring GATA boxes to which GATA1 binds highly but GATA2 binds only weakly are important for the mature erythroid cell function. Our results thus support the contention that preceding binding of GATA2 helps the following binding of GATA1 and thereby secures smooth expression of the transient-phase genes.


Assuntos
Células Eritroides/citologia , Eritropoese/fisiologia , Fator de Transcrição GATA1/genética , Fator de Transcrição GATA2/genética , Células-Tronco Hematopoéticas/citologia , Animais , Sítios de Ligação , Diferenciação Celular , Células Eritroides/metabolismo , Fator de Transcrição GATA1/metabolismo , Fator de Transcrição GATA2/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Hematopoéticas/metabolismo , Camundongos , Camundongos Transgênicos
18.
Genes Cells ; 18(10): 886-98, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23890289

RESUMO

GATA1 is a transcription factor essential for erythropoiesis and megakaryopoiesis. It has been found that Gata1 gene knockdown heterozygous female (Gata1(G1.05/+)) mice spontaneously develop erythroblastic leukemias. In this study, we have generated a novel Gata1 knockdown erythroblastic cell line, designated GAK14, from the leukemia cells in the Gata1(G1.05/+) mice. Although GAK14 cells maintain immature phenotype on OP9 stromal cells in the presence of erythropoietin and stem cell factor, the cells produce Gr-1-, Mac1-, B220-, CD3e- or CD49b-positive hematopoietic cells when co-cultured with DAS104-8 feeder cells. However, GAK14 cells did not produce erythroid and megakaryocytic lineages, perhaps due to the absence of GATA1. Indeed, GAK14 cells became capable of differentiating into mature erythroid cells when complemented with full-length GATA1 and co-cultured with fetal liver-derived FLS5 stromal cells. This differentiation potential was impaired when GATA1 lacking the N-terminal domain was complemented. The N-terminal domain is known to contribute to the pathogenesis of transient abnormal myelopoiesis and acute megakaryoblastic leukemia related to Down syndrome. These results thus showed that GAK14 cells will serve as a powerful tool for dissecting domain function of GATA1 and that the GATA1 N-terminal domain is essential for the erythroid differentiation of GAK14 cells.


Assuntos
Aminoácidos/genética , Linhagem Celular Tumoral , Células Precursoras Eritroides/fisiologia , Eritropoese , Fator de Transcrição GATA1/química , Fator de Transcrição GATA1/metabolismo , Leucemia Eritroblástica Aguda , Animais , Linhagem da Célula , Técnicas de Cocultura , Células Precursoras Eritroides/metabolismo , Feminino , Fator de Transcrição GATA1/genética , Expressão Gênica , Técnicas de Silenciamento de Genes , Células Progenitoras de Megacariócitos/fisiologia , Camundongos , Mielopoese , Estrutura Terciária de Proteína
19.
Cell Struct Funct ; 38(2): 155-67, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23748603

RESUMO

The mina53 (myc-induced nuclear antigen with a 53 kDa molecular mass; also known as mina) was identified as a direct transcriptional target of the oncoprotein Myc and encodes a conserved protein in vertebrates. While Mina53 is known to be associated with tumorigenesis, it is not clear what role Mina53 plays in non-neoplastic tissues. To directly address the roles of Mina53 in non-neoplastic tissues, we created mina53-deficient mice. Both male and female mina53-deficient mice reached adulthood and were fertile, suggesting that Mina53 is dispensable for the basic developmental processes. Since we found that Mina53 was expressed in cells responsible for immune responses, we investigated whether Mina53 was involved in immune responses. When mice were exposed intranasally to house dust mites as an allergen, the airway tract showed hyperresponsiveness to methacholine in wild-type mice but not in mina53-deficient mice. The mina53-deficient mice also showed a significantly reduced migration of immune cells, including eosinophils, into bronchoalveolar lavage fluid compared with wild-type mice. The levels of Th2 cytokines, IL-4 and IL-5, produced in response to house dust mites were lower in the mina53-deficient mice than in wild-type mice. The level of IFN-γ in bronchoalveolar lavage fluid was significantly decreased by exposure to house dust mites in wild-type mice but not in the mina53-deficient mice. These results suggest that Mina53 plays a role in the allergic response to inhaled allergens, possibly through controlling IL-4 production.


Assuntos
Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Hipersensibilidade Respiratória/imunologia , Alérgenos/imunologia , Animais , Líquido da Lavagem Broncoalveolar/citologia , Movimento Celular/imunologia , Eosinófilos/imunologia , Feminino , Células Caliciformes/imunologia , Imunoglobulina E/sangue , Interferon gama/metabolismo , Interleucina-4/metabolismo , Interleucina-5/metabolismo , Macrófagos/imunologia , Masculino , Cloreto de Metacolina/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ácaros/imunologia , Proteínas de Neoplasias/deficiência , Proteínas Nucleares/deficiência
20.
Mol Cell Biol ; 32(23): 4742-55, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22988301

RESUMO

The zinc finger transcription factors GATA1 and GATA2 participate in mast cell development. Although the expression of these factors is regulated in a cell lineage-specific and differentiation stage-specific manner, their regulation during mast cell development has not been clarified. Here, we show that the GATA2 mRNA level was significantly increased while GATA1 was maintained at low levels during the differentiation of mast cells derived from mouse bone marrow (BMMCs). Unlike in erythroid cells, forced expression or small interfering RNA (siRNA)-mediated knockdown of GATA1 rarely affected GATA2 expression, and vice versa, in mast cells, indicating the absence of cross-regulation between Gata1 and Gata2 genes. Chromatin immunoprecipitation assays revealed that both GATA factors bound to most of the conserved GATA sites of Gata1 and Gata2 loci in BMMCs. However, the GATA1 hematopoietic enhancer (G1HE) of the Gata1 gene, which is essential for GATA1 expression in erythroid and megakaryocytic lineages, was bound only weakly by both GATA factors in BMMCs. Furthermore, transgenic-mouse reporter assays revealed that the G1HE is not essential for reporter expression in BMMCs and peritoneal mast cells. Collectively, these results demonstrate that the expression of GATA factors in mast cells is regulated in a manner quite distinct from that in erythroid cells.


Assuntos
Diferenciação Celular , Células Eritroides/metabolismo , Fator de Transcrição GATA1/genética , Fator de Transcrição GATA2/genética , Regulação da Expressão Gênica no Desenvolvimento , Mastócitos/metabolismo , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Linhagem da Célula , Células Cultivadas , Células Eritroides/citologia , Fator de Transcrição GATA1/metabolismo , Fator de Transcrição GATA2/metabolismo , Histonas/genética , Histonas/metabolismo , Masculino , Mastócitos/citologia , Camundongos , Camundongos Knockout , Ligação Proteica , RNA Mensageiro/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA