Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Development ; 147(2)2020 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-31852686

RESUMO

Lymphangiogenesis plays important roles in normal fetal development and postnatal growth. However, its molecular regulation remains unclear. Here, we have examined the function of forkhead box protein O1 (FOXO1) transcription factor, a known angiogenic factor, in developmental dermal lymphangiogenesis using endothelial cell-specific FOXO1-deficient mice. FOXO1-deficient mice showed disconnected and dilated lymphatic vessels accompanied with increased proliferation and decreased apoptosis in the lymphatic capillaries. Comprehensive DNA microarray analysis of the causes of in vivo phenotypes in FOXO1-deficient mice revealed that the gene encoding C-X-C chemokine receptor 4 (CXCR4) was the most drastically downregulated in FOXO1-deficient primary lymphatic endothelial cells (LECs). CXCR4 was expressed in developing dermal lymphatic capillaries in wild-type mice but not in FOXO1-deficient dermal lymphatic capillaries. Furthermore, FOXO1 suppression impaired migration toward the exogenous CXCR4 ligand, C-X-C chemokine ligand 12 (CXCL12), and coordinated proliferation in LECs. These results suggest that FOXO1 serves an essential role in normal developmental lymphangiogenesis by promoting LEC migration toward CXCL12 and by regulating their proliferative activity. This study provides valuable insights into the molecular mechanisms underlying developmental lymphangiogenesis.


Assuntos
Derme/metabolismo , Proteína Forkhead Box O1/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Linfangiogênese/genética , Receptores CXCR4/genética , Cauda/metabolismo , Regulação para Cima/genética , Animais , Animais Recém-Nascidos , Antígenos CD/metabolismo , Apoptose , Sequência de Bases , Caderinas/metabolismo , Morte Celular , Proliferação de Células , Quimiocina CXCL12/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Elementos Facilitadores Genéticos/genética , Deleção de Genes , Integrases/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Regiões Promotoras Genéticas/genética , Ligação Proteica , Receptores CXCR4/metabolismo
2.
Biochem Biophys Res Commun ; 556: 134-141, 2021 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-33839409

RESUMO

Oxidative stress is a deteriorating factor for pancreatic ß-cells under chronic hyperglycemia in diabetes. However, the molecular mechanism underlying the increase in oxidative stress in ß-cells under diabetic conditions remains unclear. We demonstrated previously that the selective alleviation of glucotoxicity ameliorated the downregulation of several ß-cell factors, including Cox6a2. Cox6a2 encodes a subunit of the respiratory chain complex IV in mitochondria. In this study, we analyzed the role of Cox6a2 in pancreatic ß-cell function and its pathophysiological significance in diabetes mellitus. Cox6a2-knockdown experiments in MIN6-CB4 cells indicated an increased production of reactive oxygen species as detected by CellROX Deep Red reagent using flow cytometry. In systemic Cox6a2-knockout mice, impaired glucose tolerance was observed under a high-fat high-sucrose diet. However, insulin resistance was reduced when compared with control littermates. This indicates a relative insufficiency of ß-cell function. To examine the transcriptional regulation of Cox6a2, ATAC-seq with islet DNA was performed and an open-chromatin area within the Cox6a2 enhancer region was detected. Reporter gene analysis using this area revealed that MafA directly regulates Cox6a2 expression. These findings suggest that the decreased expression of Cox6a2 increases the levels of reactive oxygen species and that Mafa is associated with decreased Cox6a2 expression under glucotoxic conditions.


Assuntos
Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Proteínas Musculares/deficiência , Espécies Reativas de Oxigênio/metabolismo , Animais , Linhagem Celular , Diabetes Mellitus Experimental/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/biossíntese , Complexo IV da Cadeia de Transporte de Elétrons/genética , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Regulação da Expressão Gênica , Glucose/metabolismo , Intolerância à Glucose/genética , Células HEK293 , Humanos , Insulina/metabolismo , Resistência à Insulina/genética , Fatores de Transcrição Maf Maior/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Proteínas Musculares/biossíntese , Proteínas Musculares/genética , Estresse Oxidativo , Transcrição Gênica
3.
J Clin Densitom ; 24(2): 319-329, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33172803

RESUMO

OBJECTIVE: The objective of this research was to develop 3D registration analysis method in longitudinal studies of high-resolution peripheral quantitative computed tomography (HR-pQCT), to analyze ranges of bone microstructure parameters in addition to standard parameters, and to test the precision of these measurements. METHODS: Scans of HR-pQCT and analysis of bone microstructure were performed at 3 times in 15 subjects. The 3 images were matched 3-dimensionally, and bone microstructures were analyzed in the common region. In addition to standard measurement parameters of geometry, bone mineral density (BMD), trabecular bone, and cortical bone, parameters showing plate to rod-like structure, connectivity, cavity formation of trabecular bone, and bending stability of cortical bone were also measured. Precision was evaluated with the root mean square percent coefficient variance (RMS%CV). RESULTS: RMS%CV was 0.1%-1.3% for geometry, 0.6%-1.9% for BMD, 0.8%-3.3% for trabecular bone, 2.1%-9.8% for additionally measured trabecular bone, 1.0%-3.4% for cortical bone excluding Ct.Po, 6.0%-6.1% for Ct.Po, and 0.8%-1.5% for additionally measured cortical bone. Precision was higher for 3D registration than for 2D registration in geometry, BV/TV, and Ct.Po. CONCLUSIONS: 3D registration analysis of a range of bone microstructural parameters in longitudinal analysis of HR-pQCT showed good precision, offering potential for contributing to future research on osteoporosis and bone metabolic diseases.


Assuntos
Densidade Óssea , Osso Esponjoso , Osso Esponjoso/diagnóstico por imagem , Osso Cortical , Humanos , Estudos Longitudinais , Tomografia Computadorizada por Raios X
4.
EMBO Rep ; 19(4)2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29437694

RESUMO

The piRNA pathway is a piRNA-guided retrotransposon silencing system which includes processing of retrotransposon transcripts by PIWI-piRNAs in secondary piRNA biogenesis. Although several proteins participate in the piRNA pathway, the ones crucial for the cleavage of target RNAs by PIWI-piRNAs have not been identified. Here, we show that GTSF1, an essential factor for retrotransposon silencing in male germ cells in mice, associates with both MILI and MIWI2, mouse PIWI proteins that function in prospermatogonia. GTSF1 deficiency leads to a severe defect in the production of secondary piRNAs, which are generated from target RNAs of PIWI-piRNAs. Furthermore, in Gtsf1 mutants, a known target RNA of PIWI-piRNAs is left unsliced at the cleavage site, and the generation of secondary piRNAs from this transcript is defective. Our findings indicate that GTSF1 is a crucial factor for the slicing of target RNAs by PIWI-piRNAs and thus affects secondary piRNA biogenesis in prospermatogonia.


Assuntos
Regulação da Expressão Gênica , Proteínas/metabolismo , RNA Interferente Pequeno/genética , Transcrição Gênica , Células-Tronco Germinativas Adultas/metabolismo , Animais , Núcleo Celular/metabolismo , Amplificação de Genes , Inativação Gênica , Genes de Partícula A Intracisternal , Peptídeos e Proteínas de Sinalização Intracelular , Elementos Nucleotídeos Longos e Dispersos , Masculino , Camundongos , Camundongos Knockout , Modelos Biológicos , Complexos Multiproteicos/metabolismo , Ligação Proteica , Transporte Proteico , Proteínas/genética , Interferência de RNA , Proteínas Recombinantes de Fusão , Retroelementos , Testículo/metabolismo
5.
Angiogenesis ; 21(2): 203-214, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29185141

RESUMO

Forkhead box protein O1 (FoxO1) is a transcription factor and a critical regulator of angiogenesis. Various environmental stimuli, including growth factors, nutrients, shear stress, oxidative stress and hypoxia, affect FoxO1 subcellular localization and strongly influence its transcriptional activity; however, FoxO1-localization patterns in endothelial cells (ECs) during development have not been clarified in vivo. Here, we reported that FoxO1 expression was observed in three layers of angiogenic vessels in developing mouse retinas and that among these layers, the front layer showed high levels of FoxO1 expression in the nuclei of most tip ECs. Because tip ECs migrate toward the avascular hypoxic area, we focused on hypoxia as a major stimulus regulating FoxO1 subcellular localization in tip cells. In cultured ECs, FoxO1 accumulated into the nucleus under hypoxic conditions, with hypoxia also inducing expression of tip-cell-specific genes, including endothelial-specific molecule 1 (ESM1), which was suppressed by FoxO1 knockdown. Additionally, in murine models, EC-specific FoxO1 deletion resulted in reduced ESM1 expression and suppressed tip-cell migration during angiogenesis. These findings indicated roles for FoxO1 in tip-cell migration and that its transcriptional activity is regulated by hypoxia.


Assuntos
Células Endoteliais/metabolismo , Proteína Forkhead Box O1/metabolismo , Regulação da Expressão Gênica , Hipóxia/metabolismo , Retina/crescimento & desenvolvimento , Neovascularização Retiniana/metabolismo , Animais , Células Endoteliais/patologia , Proteína Forkhead Box O1/genética , Técnicas de Silenciamento de Genes , Humanos , Hipóxia/genética , Hipóxia/patologia , Camundongos , Camundongos Transgênicos , Retina/patologia , Neovascularização Retiniana/genética , Neovascularização Retiniana/patologia
6.
Genes Cells ; 17(9): 758-67, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22845550

RESUMO

Several reports have suggested that Foxo1, a key regulator in differentiation, growth and metabolism, is involved in pancreatic ß-cell function. However, detailed analyses have been hampered by a lack of Foxo1-deficient ß cells. To elucidate Foxo1's function in ß cells, we produced a ß-cell line with inducible Foxo1 deletion. We generated a conditional knockout mouse line, in which Cre recombinase deletes the Foxo1 gene. We then established a ß-cell line from an insulinoma induced in this knockout mouse by the ß-cell-specific expression of simian virus 40 T antigen. In this cell line, designated MIN6-Foxo1flox/flox, adenovirus-mediated Cre expression ablates the Foxo1 gene, generating MIN6-Foxo1-KO cells. Using these knockout and floxed cell lines, we found that Foxo1 ablation enhanced the glucose-stimulated insulin secretion (GSIS) at high glucose concentrations and enhanced ß-cell proliferation. We also conducted DNA microarray analyses of MIN6-Foxo1-KO cells infected with either an adenovirus vector expressing a constitutively active FOXO1 or a control vector and identified several Foxo1-regulated genes, including some known to be related to ß-cell function. These cells should be useful for further studies on Foxo1's roles in ß-cells and may lead to novel strategies for treating the impaired insulin secretion in type 2 diabetes mellitus.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Alelos , Animais , Antígenos Transformantes de Poliomavirus/genética , Antígenos Transformantes de Poliomavirus/metabolismo , Apoptose , Linhagem Celular , Proliferação de Células , Quimera/genética , Quimera/metabolismo , Feminino , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Glucose/farmacologia , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Integrases/genética , Integrases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Vírus 40 dos Símios/genética , Vírus 40 dos Símios/metabolismo
7.
Commun Biol ; 6(1): 771, 2023 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-37488353

RESUMO

The Zfp296 gene encodes a zinc finger-type protein. Its expression is high in mouse embryonic stem cells (ESCs) but rapidly decreases following differentiation. Zfp296-knockout (KO) ESCs grew as flat colonies, which were reverted to rounded colonies by exogenous expression of Zfp296. KO ESCs could not form teratomas when transplanted into mice but could efficiently contribute to germline-competent chimeric mice following blastocyst injection. Transcriptome analysis revealed that Zfp296 deficiency up- and down-regulates a distinct group of genes, among which Dppa3, Otx2, and Pou3f1 were markedly downregulated. Chromatin immunoprecipitation sequencing demonstrated that ZFP296 binding is predominantly seen in the vicinity of the transcription start sites (TSSs) of a number of genes, and ZFP296 was suggested to negatively regulate transcription. Consistently, chromatin accessibility assay clearly showed that ZFP296 binding reduces the accessibility of the TSS regions of target genes. Zfp296-KO ESCs showed increased histone H3K9 di- and trimethylation. Co-immunoprecipitation analyses revealed interaction of ZFP296 with G9a and GLP. These results show that ZFP296 plays essential roles in maintaining the global epigenetic state of ESCs through multiple mechanisms including activation of Dppa3, attenuation of chromatin accessibility, and repression of H3K9 methylation, but that Zfp296-KO ESCs retain a unique state of pluripotency while lacking the teratoma-forming ability.


Assuntos
Cromatina , Teratoma , Animais , Camundongos , Células-Tronco Embrionárias , Histonas , Células-Tronco Embrionárias Murinas , Bioensaio , Proteínas Cromossômicas não Histona , Fator 6 de Transcrição de Octâmero
8.
Bone ; 160: 116416, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35398293

RESUMO

PURPOSE: The effects of daily teriparatide (20 µg) (D-PTH), weekly high-dose teriparatide (56.5 µg) (W-PTH), or bisphosphonates (BPs) on areal bone mineral density (aBMD), bone turnover markers (BTMs), volumetric BMD (vBMD), microarchitecture, and estimated strength were investigated in postmenopausal osteoporosis patients. METHODS: The study participants were 131 women with a history of fragility fractures. They were randomized to receive D-PTH, W-PTH, or BPs (alendronate or risedronate) for 18 months. Dual-energy X-ray absorptiometry (DXA), BTMs, and high-resolution peripheral quantitative CT (HR-pQCT) parameters were evaluated at baseline and after 6 and 18 months of treatment. The primary endpoint was the change (%) in cortical thickness (Ct.Th) after 18 months' treatment compared with baseline. RESULTS: DXA showed that D-PTH, W-PTH, and BPs increased lumbar spine aBMD (+12.0%, +8.5%, and +6.8%) and total hip aBMD (+3.0%, +2.1%, and +3.0%), but D-PTH and W-PTH decreased 1/3 radius aBMD (-4.1%, -3.0%, -1.4%) after 18 months. On HR-pQCT, D-PTH increased trabecular vBMD (Tb.vBMD) at the distal radius and tibia after 18 months (+6.4%, +3.7%) compared with the BPs group, decreased cortical volumetric tissue mineral density (Ct.vTMD) (-1.8%, -0.9%) compared with the other groups, increased Ct.Th (+1.3%, +3.9%), and increased failure load (FL) (+4.7%, +4.4%). W-PTH increased Tb.vBMD (+5.3%, +1.9%), maintained Ct.vTMD (-0.7%, +0.2%) compared with D-PTH, increased Ct.Th (+0.6%, +3.6%), and increased FL (+4.9%, +4.5%). The BPs increased Tb.vBMD only in the radius (+2.0%, +0.2%), maintained Ct.vTMD (-0.6%, +0.3%), increased Ct.Th (+0.5%, +3.4%), and increased FL (+3.9%, +2.8%). CONCLUSIONS: D-PTH and W-PTH comparably increased Ct.Th, the primary endpoint. D-PTH had a strong effect on trabecular bone. Although D-PTH decreased Ct.vTMD, it increased Ct.Th and total bone strength. W-PTH had a moderate effect on trabecular bone, maintained Ct.vTMD, and increased Ct.Th and total bone strength to the same extent as D-PTH.


Assuntos
Osteoporose Pós-Menopausa , Teriparatida , Absorciometria de Fóton , Densidade Óssea , Difosfonatos/farmacologia , Difosfonatos/uso terapêutico , Feminino , Humanos , Osteoporose Pós-Menopausa/diagnóstico por imagem , Osteoporose Pós-Menopausa/tratamento farmacológico , Rádio (Anatomia)/diagnóstico por imagem , Teriparatida/farmacologia , Teriparatida/uso terapêutico , Tíbia
9.
Genes Cells ; 15(8): 813-28, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20590823

RESUMO

In a search for genes specifically expressed in mouse embryonic stem cells, we identified one we called Ces5. We found that it corresponded to the Ooep gene, which was recently reported to be expressed specifically in oocytes. Mouse Ces5/Ooep, also called Moep19 or Floped, encoded a 164-amino acid protein, which was detected in the cytoplasm of developing and mature oocytes and in embryos throughout the preimplantation period. To examine its function, we carried out targeted disruption of this gene. The Ces5/Ooep-null mice were grossly normal, but the females were infertile. Although the ovaries and ovulation appeared normal, the embryos from Ces5/Ooep-null females mated with wild-type males showed developmental arrest at the two- or four-cell stage. In addition, their first cleavage was considerably delayed and often asymmetrical. Thus, Ces5/Ooep is a maternal-effect gene. By electron microscopy, we found that the eggs from Ces5/Ooep-null females lacked oocyte cytoplasmic lattices (CPLs), which have long been predicted to function as a storage form for components that are maternally contributed to the early embryo. Further analysis showed that CES5/OOEP was directly associated with the CPLs. These results indicate that CES5/OOEP is an essential component of the CPLs and is required for embryonic development at the maternal-zygotic stage transition.


Assuntos
Citoplasma/metabolismo , Embrião de Mamíferos/embriologia , Oócitos/citologia , Oócitos/metabolismo , Proteínas de Ligação a RNA/metabolismo , Zigoto/metabolismo , Sequência de Aminoácidos , Animais , Embrião de Mamíferos/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Masculino , Camundongos , Camundongos Knockout , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/genética , Transcrição Gênica/genética , Zigoto/citologia
10.
Sci Rep ; 11(1): 477, 2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33436850

RESUMO

A pancreatic ß-cell line MIN6 was previously established in our lab from an insulinoma developed in an IT6 transgenic mouse expressing the SV40 T antigen in ß-cells. This cell line has been widely used for in vitro analysis of ß-cell function, but tends to lose the mature ß-cell features, including glucose-stimulated insulin secretion (GSIS), in long-term culture. The aim of this study was to develop a stable ß-cell line that retains the characteristics of mature ß-cells. Considering that mice derived from a cross between C3H and C57BL/6 strains are known to exhibit higher insulin secretory capacity than C57BL/6 mice, an IT6 male mouse of this hybrid background was used to isolate insulinomas, which were independently cultured. After 7 months of continuous culturing, we obtained the MIN6-CB4 ß-cell line, which stably maintains its GSIS. It has been noted that ß-cell lines express the glucagon (Gcg) gene at certain levels. MIN6-CB4 cells were utilized to assess the effects of differential Gcg expression on ß-cell function. Our data show the functional importance of Gcg expression and resulting basal activation of the GLP-1 receptor in ß-cells. MIN6-CB4 cells can serve as an invaluable tool for studying the regulatory mechanisms of insulin secretion, such as the GLP-1/cAMP signaling, in ß-cells.


Assuntos
Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Glucagon/fisiologia , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Insulinoma/metabolismo , Neoplasias Pancreáticas/metabolismo , Animais , Feminino , Células Secretoras de Insulina/citologia , Insulinoma/patologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Pancreáticas/patologia
11.
Bone ; 144: 115770, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33249321

RESUMO

PURPOSE: To investigate the effects of sequential therapy with monthly intravenous ibandronate on bone mineral density (BMD) and microstructure in patients with primary osteoporosis who received teriparatide treatment. METHODS: Sixty-six patients with primary osteoporosis who had undergone teriparatide treatment for more than 12 months (mean 18.6 months) received sequential therapy with 1 mg/month intravenous ibandronate for 12 months. The patients were evaluated using dual-energy X-ray absorptiometry (DXA), quantitative ultrasound, bone turnover markers, and high-resolution peripheral quantitative computed tomography (HR-pQCT) at baseline and 6 and 12 months after beginning administration. RESULTS: At 12 months after beginning sequential therapy, the bone resorption marker, tartrate-resistant acid phosphatase-5b, decreased by 39.5%, with 82.3% of the patients exhibiting levels within the normal limit. DXA revealed that the BMD of the lumbar spine increased by 3.2%, with 79.0% of the patients exhibiting a response, and 40.3% experiencing an increase in BMD over 5%. HR-pQCT revealed that the cortical thickness of the distal tibia was increased by 2.6%. The cortical area increased by 2.5%, and the buckling ratio (an index of cortical instability) decreased by 2.5%. Most parameters of the trabecular bone showed no significant changes. These changes in the cortical bone were observed in both the distal radius and tibia and appeared beginning 6 months after treatment initiation. CONCLUSIONS: Sequential therapy with monthly intravenous ibandronate increased the BMD and improved the cortical bone microstructure of osteoporotic patients who had undergone teriparatide treatment.


Assuntos
Conservadores da Densidade Óssea , Osteoporose , Absorciometria de Fóton , Densidade Óssea , Conservadores da Densidade Óssea/uso terapêutico , Humanos , Ácido Ibandrônico , Osteoporose/diagnóstico por imagem , Osteoporose/tratamento farmacológico , Teriparatida/uso terapêutico
12.
Dev Biol ; 335(1): 216-27, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19735653

RESUMO

We recently reported that the Gtsf1/Cue110 gene, a member of the evolutionarily conserved UPF0224 family, is expressed predominantly in male germ cells, and that the GTSF1/CUE110 protein is localized to the cytoplasm of these cells in the adult testis. Here, to analyze the roles of the Gtsf1/Cue110 gene in spermatogenesis, we produced Gtsf1/Cue110-null mice by gene targeting. The Gtsf1/Cue110-null mice grew normally and appeared healthy; however, the males were sterile due to massive apoptotic death of their germ cells after postnatal day 14. In contrast, the null females were fertile. Detailed analyses revealed that the Gtsf1/Cue110-null male meiocytes ceased meiotic progression before the zygotene stage. Thus, the Gtsf1/Cue110 gene is essential for spermatogenesis beyond the early meiotic phase. Furthermore, the loss of the Gtsf1/Cue110 gene caused increased transcription of the long interspersed nucleotide element (Line-1) and the intracisternal A-particle (IAP) retrotransposons, accompanied by demethylation of their promoter regions. These observations indicate that Gtsf1/Cue110 is required for spermatogenesis and involved in retrotransposon suppression in male germ cells.


Assuntos
Proteínas , Retroelementos , Espermatogênese/fisiologia , Testículo , Dedos de Zinco , Animais , Metilação de DNA , Feminino , Fertilidade/fisiologia , Regulação da Expressão Gênica , Marcação de Genes , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Meiose/fisiologia , Camundongos , Camundongos Knockout , Proteínas/genética , Proteínas/metabolismo , Testículo/citologia , Testículo/fisiologia
13.
Dev Biol ; 325(1): 238-48, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19014927

RESUMO

The differentiation programs of spermatogenesis and oogenesis are largely independent. In the early stages, however, the mechanisms partly overlap. Here we demonstrated that a germ-cell-specific basic helix-loop-helix (bHLH) transcription factor gene, Sohlh2, is required for early spermatogenesis and oogenesis. SOHLH2 was expressed in mouse spermatogonia from the undifferentiated stage through differentiation and in primordial-to-primary oocytes. Sohlh2-null mice, produced by gene targeting, showed both male and female sterility, owing to the disrupted differentiation of mature (KIT(+)) spermatogonia and oocytes. The Sohlh2-null mice also showed the downregulation of genes involved in spermatogenesis and oogenesis, including the Sohlh1 gene, which is essential for these processes. Furthermore, we showed that SOHLH2 and SOHLH1 could form heterodimers. These observations suggested that SOHLH2 might coordinate with SOHLH1 to control spermatogonial and oocyte genes, including Sohlh1, to promote the differentiation of KIT(+) germ cells in vivo. This study lays the foundation for further dissection of the bHLH network that regulates early spermatogenesis and oogenesis.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Oócitos/citologia , Oócitos/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Espermatogônias/citologia , Espermatogônias/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Western Blotting , Linhagem Celular , Feminino , Imunofluorescência , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Masculino , Camundongos , Oogênese/genética , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espermatogênese/genética , Testículo/citologia , Testículo/metabolismo
14.
Dev Growth Differ ; 50(6): 479-83, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18445065

RESUMO

Naked plasmid DNA injected into skeletal muscle is taken up by muscle cells and the genes in the plasmid are expressed. Among the non-viral techniques for gene transfer in vivo, this method is especially simple, inexpensive, and safe. However, the relatively low expression levels attained by this method have limited its applications for uses other than as a DNA vaccine. We and other groups investigated the applicability of in vivo electroporation for gene transfer into muscle, using plasmid DNA vector. The results demonstrated that gene transfer into muscle by in vivo electroporation is far more efficient than simple intramuscular DNA injection and provides a potential approach to systemically delivering cytokines, growth factors, and other serum proteins for basic research and human gene therapy.


Assuntos
Biologia do Desenvolvimento/métodos , Eletroporação/métodos , Técnicas de Transferência de Genes , Plasmídeos/metabolismo , Animais , Citocinas/metabolismo , Feminino , Vetores Genéticos , Interleucina-5/biossíntese , Óperon Lac , Camundongos , Camundongos Endogâmicos C57BL , Músculos/metabolismo , Vacinas de DNA/química , beta-Galactosidase/metabolismo
15.
Gene Expr Patterns ; 8(1): 27-35, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17919994

RESUMO

The large number of expressed sequence tags (ESTs) now available in databases has enabled the analysis of gene expression profiles in silico. We searched public databases for uncharacterized transcripts specifically expressed in germ cells, in an attempt to identify genes involved in gametogenesis. We found a transcript that is expressed in unfertilized eggs, ovaries, and testes of the mouse. It has an open reading frame (ORF) encoding a 167-amino acid protein belonging to the UPF0224 (unknown protein family 0224) family. We called the novel gene Cue110. We examined the Pfam database for other members of the UPF0224 family, and found a conserved N-terminal portion among members of various species. To study the cellular localization of the Cue110 transcript and protein, we performed in situ hybridization and immunohistochemical analysis of the adult mouse ovary and testis. In the testis, specific hybridization signals were observed weakly in preleptotene spermatocytes but maximally in late round spermatids. Immunostaining showed that Cue110 protein was present predominantly in the cytoplasm of pachytene spermatocytes and round spermatids. In the ovary, weak hybridization signals were observed in primary oocytes in the primordial, primary, and secondary follicles, but Cue110 protein was not detected in oocytes by immunostaining. We next examined the developmental expression pattern of the Cue110 gene using RT-PCR and western blotting, and found its increasing expression coincided with the appearance of spermatocytes. Thus, the Cue110 gene is expressed predominantly in male germ cells at stages from the pachytene spermatocytes to round spermatids.


Assuntos
Gametogênese/genética , Células Germinativas/química , Proteínas/genética , Espermatócitos/química , Animais , Bases de Dados de Ácidos Nucleicos , Feminino , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Ovário/química , Proteínas/análise , RNA Mensageiro/análise , Testículo/química
16.
Diabetes Res Clin Pract ; 77 Suppl 1: S138-42, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17451835

RESUMO

Embryonic stem (ES) cells can differentiate into many cell types. Recent reports have shown that ES cells can differentiate into insulin-producing cells. We have established an ES cell line in which exogenous Pdx-1 expression was precisely regulated by the Tet-off system integrated into the ROSA26 locus and succeeded to produce insulin-producing cells. The Pdx-1 expressing final differentiated insulin-positive cells can be maintained for more than 2 months. However, in spite of their induced expression of Pdx-1, the repeated passages of cells lost their capacity to express insulin and NeuroD1 gene. Forced expression of NeuroD1 gene by adenoviral vector in these cells restored the expression of insulin. These results suggested that maintenance of the property of insulin-producing cells derived from ES cells could be achieved by synergistic expression of Pdx-1 and NeuroD1.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Regulação da Expressão Gênica , Proteínas de Homeodomínio/genética , Insulina/genética , Transativadores/genética , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Humanos , Camundongos , RNA/genética , RNA/isolamento & purificação , Transfecção
17.
Sci Rep ; 7(1): 12462, 2017 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-28963472

RESUMO

The Cys2/His2-type zinc finger protein Zfp296 has been implicated in stem cell pluripotency and tumor pathogenesis. However, its mechanisms remain elusive. Here, we demonstrated that a Zfp296 deficiency in mice impairs germ-cell development and embryonic growth. Zfp296 was intracellularly localized to heterochromatin in embryos. A GST-Zfp296 pull-down experiment using ES cell nuclear extract followed by LC-MS/MS showed that Zfp296 interacts with component proteins of heterochromatin (such as HP1, Dnmt1, Dnmt3b, and ATRX) and the NuRD complex. We focused on H3K9 methylation as a hallmark of heterochromatin, and found that Zfp296 overexpression in cultured cells reduces the Suv39h1-mediated H3K9 methylation. Consistent with this finding, in Zfp296 -/- mouse embryos, we observed a global increase in H3K9 methylation in a developmental stage-dependent manner, and showed, by ChIP-qPCR, that the H3K9me3 levels at major satellite repeats were elevated in Zfp296 -/- embryos. Our results demonstrate that Zfp296 is a component of heterochromatin that affects embryonic development by negatively regulating H3K9 methylation.


Assuntos
Proteínas de Ligação a DNA/genética , Desenvolvimento Embrionário/genética , Heterocromatina/metabolismo , Histonas/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , Processamento de Proteína Pós-Traducional , Animais , Diferenciação Celular , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , DNA (Citosina-5-)-Metiltransferase 1/genética , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Proteínas de Ligação a DNA/deficiência , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Heterocromatina/química , Histonas/genética , Masculino , Metilação , Metiltransferases/genética , Metiltransferases/metabolismo , Camundongos , Camundongos Knockout , Células-Tronco Embrionárias Murinas/citologia , Ovário/anormalidades , Ovário/crescimento & desenvolvimento , Ovário/metabolismo , Cultura Primária de Células , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Testículo/anormalidades , Testículo/crescimento & desenvolvimento , Testículo/metabolismo , Proteína Nuclear Ligada ao X/genética , Proteína Nuclear Ligada ao X/metabolismo , DNA Metiltransferase 3B
19.
PLoS One ; 11(8): e0161190, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27526291

RESUMO

A promising approach to new diabetes therapies is to generate ß cells from other differentiated pancreatic cells in vivo. Because the acinar cells represent the most abundant cell type in the pancreas, an attractive possibility is to reprogram acinar cells into ß cells. The transcription factor Pdx1 (Pancreas/duodenum homeobox protein 1) is essential for pancreatic development and cell lineage determination. Our objective is to examine whether exogenous expression of Pdx1 in acinar cells of adult mice might induce reprogramming of acinar cells into ß cells. We established a transgenic mouse line in which Pdx1 and EGFP (enhanced green fluorescent protein) could be inducibly expressed in the acinar cells. After induction of Pdx1, we followed the acinar cells for their expression of exocrine and endocrine markers using cell-lineage tracing with EGFP. The acinar cell-specific expression of Pdx1 in adult mice reprogrammed the acinar cells as endocrine precursor cells, which migrated into the pancreatic islets and differentiated into insulin-, somatostatin-, or PP (pancreatic polypeptide)-producing endocrine cells, but not into glucagon-producing cells. When the mice undergoing such pancreatic reprogramming were treated with streptozotocin (STZ), the newly generated insulin-producing cells were able to ameliorate STZ-induced diabetes. This paradigm of in vivo reprogramming indicates that acinar cells hold promise as a source for new islet cells in regenerative therapies for diabetes.


Assuntos
Células Acinares/citologia , Transdiferenciação Celular/genética , Pâncreas/citologia , Transativadores/genética , Células Acinares/metabolismo , Animais , Glicemia/metabolismo , Reprogramação Celular , Glândulas Exócrinas/citologia , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Insulina/metabolismo , Camundongos , Camundongos Transgênicos
20.
PLoS One ; 11(3): e0150390, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26930067

RESUMO

The unknown protein family 0224 (UPF0224) includes three members that are expressed in germ-line cells in mice: Gtsf1, Gtsf1l, and BC048502 (Gtsf2). These genes produce proteins with two repeats of the CHHC Zn-finger domain, a predicted RNA-binding motif, in the N terminus. We previously reported that Gtsf1 is essential for spermatogenesis and retrotransposon suppression. In this study, we investigated the expression patterns and functions of Gtsf1l and Gtsf2. Interestingly, Gtsf1l and Gtsf2 were found to be sequentially but not simultaneously expressed in gonocytes and spermatids. Pull-down experiments showed that both GTSF1L and GTSF2 can interact with PIWI-protein complexes. Nevertheless, knocking out Gtsf1, Gtsf2, or both did not cause defects in spermatogenesis or retrotransposon suppression in mice.


Assuntos
Proteínas de Transporte/genética , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/metabolismo , Proteínas/genética , Espermátides/metabolismo , Espermatogênese/genética , Sequência de Aminoácidos , Animais , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Proteínas de Transporte/metabolismo , Linhagem Celular , Células Cultivadas , Feminino , Imunofluorescência , Células Germinativas/crescimento & desenvolvimento , Immunoblotting , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Ligação Proteica , Proteínas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Espermátides/crescimento & desenvolvimento , Testículo/citologia , Testículo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA