Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 166(5): 1147-1162.e15, 2016 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-27565344

RESUMO

Alternative splicing is prevalent in the mammalian brain. To interrogate the functional role of alternative splicing in neural development, we analyzed purified neural progenitor cells (NPCs) and neurons from developing cerebral cortices, revealing hundreds of differentially spliced exons that preferentially alter key protein domains-especially in cytoskeletal proteins-and can harbor disease-causing mutations. We show that Ptbp1 and Rbfox proteins antagonistically govern the NPC-to-neuron transition by regulating neuron-specific exons. Whereas Ptbp1 maintains apical progenitors partly through suppressing a poison exon of Flna in NPCs, Rbfox proteins promote neuronal differentiation by switching Ninein from a centrosomal splice form in NPCs to a non-centrosomal isoform in neurons. We further uncover an intronic human mutation within a PTBP1-binding site that disrupts normal skipping of the FLNA poison exon in NPCs and causes a brain-specific malformation. Our study indicates that dynamic control of alternative splicing governs cell fate in cerebral cortical development.


Assuntos
Processamento Alternativo , Córtex Cerebral/embriologia , Células-Tronco Neurais/citologia , Neurogênese/genética , Neurônios/citologia , Animais , Centrossomo/metabolismo , Córtex Cerebral/anormalidades , Córtex Cerebral/citologia , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Éxons , Ribonucleoproteínas Nucleares Heterogêneas/genética , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Humanos , Camundongos , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Domínios Proteicos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Fatores de Processamento de RNA
2.
Proc Natl Acad Sci U S A ; 118(43)2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34663724

RESUMO

Although it is held that proinflammatory changes precede the onset of breast cancer, the underlying mechanisms remain obscure. Here, we demonstrate that FRS2ß, an adaptor protein expressed in a small subset of epithelial cells, triggers the proinflammatory changes that induce stroma in premalignant mammary tissues and is responsible for the disease onset. FRS2ß deficiency in mouse mammary tumor virus (MMTV)-ErbB2 mice markedly attenuated tumorigenesis. Importantly, tumor cells derived from MMTV-ErbB2 mice failed to generate tumors when grafted in the FRS2ß-deficient premalignant tissues. We found that colocalization of FRS2ß and the NEMO subunit of the IκB kinase complex in early endosomes led to activation of nuclear factor-κB (NF-κB), a master regulator of inflammation. Moreover, inhibition of the activities of the NF-κB-induced cytokines, CXC chemokine ligand 12 and insulin-like growth factor 1, abrogated tumorigenesis. Human breast cancer tissues that express higher levels of FRS2ß contain more stroma. The elucidation of the FRS2ß-NF-κB axis uncovers a molecular link between the proinflammatory changes and the disease onset.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Neoplasias da Mama/etiologia , Neoplasias da Mama/metabolismo , Neoplasias Mamárias Experimentais/etiologia , Neoplasias Mamárias Experimentais/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Animais , Neoplasias da Mama/imunologia , Carcinogênese , Citocinas/metabolismo , Feminino , Humanos , Inflamação/etiologia , Inflamação/metabolismo , Neoplasias Mamárias Experimentais/imunologia , Vírus do Tumor Mamário do Camundongo , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Gravidez , Receptor ErbB-2/metabolismo , Infecções por Retroviridae , Microambiente Tumoral/imunologia , Infecções Tumorais por Vírus
3.
Mol Psychiatry ; 27(3): 1694-1703, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34997193

RESUMO

The amygdala, a critical brain region responsible for emotional behavior, is crucially involved in the regulation of the effects of stress on emotional behavior. In the mammalian forebrain, gastrin-releasing peptide (GRP), a 27-amino-acid mammalian neuropeptide, which is a homolog of the 14-amino-acid amidated amphibian peptide bombesin, is highly expressed in the amygdala. The levels of GRP are markedly increased in the amygdala after acute stress; therefore, it is known as a stress-activated modulator. To determine the role of GRP in emotional behavior under stress, we conducted some behavioral and biochemical experiments with GRP-knockout (KO) mice. GRP-KO mice exhibited a longer freezing response than wild-type (WT) littermates in both contextual and auditory fear (also known as threat) conditioning tests only when they were subjected to acute restraint stress 20 min before the conditioning. To identify the critical neural circuits associated with the regulation of emotional memory by GRP, we conducted Arc/Arg3.1-reporter mapping in the amygdala with an Arc-Venus reporter transgenic mouse line. In the amygdalostriatal transition area (AST) and the lateral side of the basal nuclei, fear conditioning after restraint stress increased neuronal activity significantly in WT mice, and GRP KO was found to negate this potentiation only in the AST. These results indicate that the GRP-activated neurons in the AST are likely to suppress excessive fear expression through the regulation of downstream circuits related to fear learning following acute stress.


Assuntos
Bombesina , Medo , Tonsila do Cerebelo/metabolismo , Animais , Bombesina/metabolismo , Bombesina/farmacologia , Condicionamento Clássico/fisiologia , Medo/fisiologia , Peptídeo Liberador de Gastrina/metabolismo , Peptídeo Liberador de Gastrina/farmacologia , Mamíferos/metabolismo , Camundongos , Camundongos Knockout
4.
Int Immunol ; 33(9): 479-490, 2021 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-34161582

RESUMO

RNase T2, a ubiquitously expressed RNase, degrades RNAs in the endosomal compartments. RNA sensors, double-stranded RNA (dsRNA)-sensing Toll-like receptor 3 (TLR3) and single-stranded RNA (ssRNA)-sensing TLR7, are localized in the endosomal compartment in mouse macrophages. We here studied the role of RNase T2 in TLR3 and TLR7 responses in macrophages. Macrophages expressed RNase T2 and a member of the RNase A family RNase 4. RNase T2 was also expressed in plasmacytoid and conventional dendritic cells. Treatment with dsRNAs or type I interferon (IFN) up-regulated expression of RNase T2 but not RNase 4. RNase T2-deficiency in macrophages up-regulated TLR3 responses but impaired TLR7 responses. Mechanistically, RNase T2 degraded both dsRNAs and ssRNAs in vitro, and its mutants showed a positive correlation between RNA degradation and the rescue of altered TLR3 and TLR7 responses. H122A and C188R RNase T2 mutations, not H69A and E118V mutations, impaired both RNA degradation and the rescue of altered TLR3 and TLR7 responses. RNase T2 in bone marrow-derived macrophages was broadly distributed from early endosomes to lysosomes, and colocalized with the internalized TLR3 ligand poly(I:C). These results suggest that RNase T2-dependent RNA degradation in endosomes/lysosomes negatively and positively regulates TLR3 and TLR7 responses, respectively, in macrophages.


Assuntos
Endorribonucleases/metabolismo , Endossomos/metabolismo , Macrófagos/metabolismo , Glicoproteínas de Membrana/metabolismo , RNA de Cadeia Dupla/metabolismo , Receptor 3 Toll-Like/metabolismo , Receptor 7 Toll-Like/metabolismo , Animais , Linhagem Celular , Citocinas/metabolismo , Células Dendríticas/metabolismo , Células HEK293 , Humanos , Lisossomos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
5.
Am J Primatol ; 84(10): e23343, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-34762313

RESUMO

Environmental enrichment is essential for the well-being of zoo animals. Recent advances in sensor and video technologies may contribute to improvements in enrichment in terms of their flexibilities and time constraints. The purpose of this study was to investigate whether interactive movie art can be used as a form of environmental enrichment. We implemented interactive movies designed by a professional artist, a visual art aiming to reflect naturalistic forest habitat, in an indoor chimpanzee enclosure at Kyoto City Zoo in Japan. Motion-tracking sensors embedded in buoys were installed at several locations around the indoor enclosure; the chimpanzees could change the movie contents by physically interacting with these objects. We recorded behaviors by observing entire troop of chimpanzees (six) between March 16 and 20, 2020 (control condition), then recorded behaviors when the interactive movie was presented (experimental condition) between March 21 and 29, 2020. Behaviors were recorded via direct observations and video recordings to examine any changes after the installation of interactive art. The chimpanzees spent more time in the indoor enclosures during the experimental condition than during the control condition. Activity budgets did not change substantially during the study period. There was no evidence of habituation to the movie during the study period. Three chimpanzees, including two young chimpanzees, interacted with the movie more frequently than the others; these young chimpanzees occasionally showed playful expressions when interacting with the movie and exhibited different reactivities to the movie scenes. These results demonstrate, first, that the interactive art did not negatively affect chimpanzee behavior, and second, that some of the chimpanzees indeed showed positive responses to the art. This study, therefore, introduces a novel possibility for environmental enrichment in zoos, involving a collaboration between science and art.


Assuntos
Animais de Zoológico , Pan troglodytes , Animais , Animais de Zoológico/fisiologia , Comportamento Animal , Florestas , Pan troglodytes/fisiologia , Projetos Piloto , Gravação em Vídeo
6.
Int Immunol ; 31(3): 157-166, 2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30476084

RESUMO

The RNA-binding protein polypyrimidine tract-binding protein-1 (Ptbp1) binds to the pyrimidine-rich sequence of target RNA and controls gene expression via post-transcriptional regulation such as alternative splicing. Although Ptbp1 is highly expressed in B lymphocytes, its role to date is largely unknown. To clarify the role of Ptbp1 in B-cell development and function, we generated B-cell-specific Ptbp1-deficient (P1BKO) mice. B-cell development in the bone marrow, spleen and peritoneal cavity of the P1BKO mice was nearly normal. However, the P1BKO mice had significantly lower levels of natural antibodies in serum compared with those of the control mice. To investigate the effect of Ptbp1 deficiency on the immune response in vivo, we immunized the P1BKO mice with T-cell-independent type-2 (TI-2) antigen NP-Ficoll and T-cell-dependent (TD) antigen NP-CGG. We found that B-cell-specific Ptbp1 deficiency causes an immunodeficiency phenotype due to defective production of antibody against both TI-2 and TD antigen. This immunodeficiency was accompanied by impaired B-cell receptor (BCR)-mediated B-cell activation and plasmablast generation. These findings demonstrate that Ptbp1 is essential for the humoral immune response.


Assuntos
Formação de Anticorpos/imunologia , Ribonucleoproteínas Nucleares Heterogêneas/imunologia , Proteína de Ligação a Regiões Ricas em Polipirimidinas/imunologia , Receptores de Antígenos de Linfócitos B/imunologia , Animais , Reações Antígeno-Anticorpo , Antígenos T-Independentes/imunologia , Linfócitos B/imunologia , Ribonucleoproteínas Nucleares Heterogêneas/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína de Ligação a Regiões Ricas em Polipirimidinas/deficiência , Linfócitos T/imunologia
7.
Immunity ; 35(1): 69-81, 2011 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-21683627

RESUMO

Toll-like receptor-7 (TLR7) and 9, innate immune sensors for microbial RNA or DNA, have been implicated in autoimmunity. Upon activation, TLR7 and 9 are transported from the endoplasmic reticulum (ER) to endolysosomes for nucleic acid sensing by an ER-resident protein, Unc93B1. Little is known, however, about a role for sensor transportation in controlling autoimmunity. TLR9 competes with TLR7 for Unc93B1-dependent trafficking and predominates over TLR7. TLR9 skewing is actively maintained by Unc93B1 and reversed to TLR7 if Unc93B1 loses preferential binding via a D34A mutation. We here demonstrate that mice harboring a D34A mutation showed TLR7-dependent, systemic lethal inflammation. CD4(+) T cells showed marked differentiation toward T helper 1 (Th1) or Th17 cell subsets. B cell depletion abolished T cell differentiation and systemic inflammation. Thus, Unc93B1 controls homeostatic TLR7 activation by balancing TLR9 to TLR7 trafficking.


Assuntos
Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Células Th1/metabolismo , Células Th17/metabolismo , Receptor 7 Toll-Like/metabolismo , Receptor Toll-Like 9/metabolismo , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos B/patologia , Diferenciação Celular , Células Cultivadas , Inflamação , Depleção Linfocítica , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mutação/genética , Ligação Proteica/genética , Transporte Proteico , Células Th1/imunologia , Células Th1/patologia , Células Th17/imunologia , Células Th17/patologia , Receptor 7 Toll-Like/genética , Receptor 7 Toll-Like/imunologia , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/imunologia
8.
J Pathol ; 249(1): 39-51, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30953353

RESUMO

CREB-binding protein (CBP) and p300 have oncogenic properties; both co-operate with pro-oncogenic transcription factors downstream of Ras-Erk signalling to support cell proliferation. By contrast, missense, truncating and in-frame mutations of CBP/p300 are found frequently in some human cancers, including cutaneous squamous cell carcinomas that originate from epidermal keratinocytes. Data support that dysfunction of CBP/p300 contributes to keratinocyte hyperproliferation and tumourigenesis; however, the mechanism by which dysfunction of CBP/p300 affects keratinocytes is unknown. Here, we used mice harbouring keratinocyte-specific genetic modifications to examine the role of CBP/p300 in the epidermis. While a single copy of either Crebbp or Ep300 was necessary and sufficient for maintaining epidermal development, reduced expression of CBP/p300 strengthened the Ras-Erk signalling-induced hyperplastic phenotype of epidermal keratinocytes. Reduced CBP/p300 expression increased ligand-induced EGFR activity while decreasing basal expression of Mig6, a negative regulator of EGFR. A reduction in CBP/p300, in combination with increased Ras-Erk signalling, also promoted epidermal tumour formation in mice. Thus, our findings support that CBP/p300 acts as a tumour suppressor in epidermal keratinocytes by counteracting EGFR-Ras-Erk signalling. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Proteína de Ligação a CREB/metabolismo , Transformação Celular Neoplásica/metabolismo , Proteína p300 Associada a E1A/metabolismo , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Queratinócitos/enzimologia , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Neoplasias Cutâneas/enzimologia , Animais , Proteína de Ligação a CREB/genética , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Células Cultivadas , Proteína p300 Associada a E1A/genética , Receptores ErbB/genética , MAP Quinases Reguladas por Sinal Extracelular/genética , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Queratinócitos/patologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Proteínas Proto-Oncogênicas p21(ras)/genética , Transdução de Sinais , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Carga Tumoral
9.
J Reprod Dev ; 65(1): 37-46, 2019 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-30416150

RESUMO

Polypyrimidine tract-binding protein 1 (PTBP1) is a highly conserved RNA-binding protein that is a well-known regulator of alternative splicing. Testicular tissue is one of the richest tissues with respect to the number of alternative splicing mRNA isoforms, but the molecular role(s) of PTBP1 in the regulation of these isoforms during spermatogenesis is still unclear. Here, we developed a germ cell-specific Ptbp1 conditional knockout (cKO) mouse model by using the Cre-loxP system to investigate the role of PTBP1 in spermatogenesis. Testis weight in Ptbp1 cKO mice was comparable to that in age-matched controls until 3 weeks of age; at ≥ 2 months old, testis weight was significantly lighter in cKO mice than in age-matched controls. Sperm count in Ptbp1 cKO mice at 2 months old was comparable to that in controls, whereas sperm count significantly decreased at 6 months old. Seminiferous tubules that exhibited degeneration in spermatogenic function were more evident in the 2-month-old Ptbp1 cKO mice than in controls. In addition, the early neonatal proliferation of spermatogonia, during postnatal days 1-5, was significantly retarded in Ptbp1 cKO mice compared with that in controls. An in vitro spermatogonia culture model (germline stem cells) revealed that hydroxytamoxifen-induced deletion of PTBP1 from germline stem cells caused severe proliferation arrest accompanied by an increase of apoptotic cell death. These data suggest that PTBP1 contributes to spermatogenesis through regulation of spermatogonia proliferation.


Assuntos
Proliferação de Células/fisiologia , Ribonucleoproteínas Nucleares Heterogêneas/fisiologia , Proteína de Ligação a Regiões Ricas em Polipirimidinas/fisiologia , Espermatogênese/fisiologia , Espermatogônias/citologia , Processamento Alternativo/fisiologia , Animais , Apoptose , Expressão Gênica , Ribonucleoproteínas Nucleares Heterogêneas/deficiência , Ribonucleoproteínas Nucleares Heterogêneas/genética , Masculino , Camundongos Knockout , Tamanho do Órgão , Proteína de Ligação a Regiões Ricas em Polipirimidinas/deficiência , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Túbulos Seminíferos/fisiologia , Contagem de Espermatozoides , Espermatócitos/metabolismo , Espermatogônias/metabolismo , Testículo/citologia , Testículo/crescimento & desenvolvimento
10.
Biochem Biophys Res Commun ; 501(3): 745-750, 2018 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-29753741

RESUMO

Hindlimb unloading (HU) of rodents has been used as a ground-based model of spaceflight. In this study, we investigated the detailed impact of 14-day HU on the murine thymus. Thymic mass and cell number were significantly reduced after 14 days of hindlimb unloading, which was accompanied by an increment of plasma corticosterone. Although corticosterone reportedly causes selective apoptosis of CD4+CD8+ thymocytes (CD4+CD8+DPs) in mice treated with short-term HU, the reduction of thymocyte cellularity after the 14-day HU was not selective for CD4+CD8+DPs. In addition to the thymocyte reduction, the cellularity of thymic epithelial cells (TECs) was also reduced by the 14-day HU. Flow cytometric and RNA-sequencing analysis suggested that medullary TECs (mTECs) were preferentially reduced after HU. Moreover, immunohistochemical staining suggested that the 14-day HU caused a reduction of the mTECs expressing autoimmune regulator (Aire). Our data suggested that HU impacts both thymocytes and TECs. Consequently, these data imply that thymic T cell repertoire formation could be disturbed during spaceflight-like stress.


Assuntos
Células Epiteliais/citologia , Elevação dos Membros Posteriores/métodos , Timócitos/citologia , Timo/fisiologia , Fatores de Transcrição/análise , Animais , Antígenos CD4/análise , Antígenos CD8/análise , Contagem de Células , Masculino , Camundongos Endogâmicos C57BL , Tamanho do Órgão , Timo/citologia , Fatores de Tempo , Proteína AIRE
11.
J Cell Sci ; 127(Pt 4): 845-57, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24357720

RESUMO

The lymphatic endothelial cell (LEC) fate decision program during development has been described. However, the mechanism underlying the maintenance of differentiated LEC identity remains largely unknown. Here, we show that fibroblast growth factor 2 (FGF2) plays a fundamental role in maintaining a differentiated LEC trait. In addition to demonstrating the appearance of LECs expressing α-smooth muscle actin in mouse lymphedematous skin in vivo, we found that mouse immortalised LECs lose their characteristics and undergo endothelial-to-mesenchymal transition (EndMT) when cultured in FGF2-depleted medium. FGF2 depletion acted synergistically with transforming growth factor (TGF) ß to induce EndMT. We also found that H-Ras-overexpressing LECs were resistant to EndMT. Activation of H-Ras not only upregulated FGF2-induced activation of the Erk mitogen activated protein kinases (MAPK3 and MAPK1), but also suppressed TGFß-induced activation of Smad2 by modulating Smad2 phosphorylation by MAPKs. These results suggest that FGF2 regulates LEC-specific gene expression and suppresses TGFß signalling in LECs through Smad2 in a Ras-MAPK-dependent manner. Taken together, our findings provide a new insight into the FGF2-Ras-MAPK-dependent mechanism that maintains and modulates the LEC trait.


Assuntos
Células Endoteliais/metabolismo , Fator 2 de Crescimento de Fibroblastos/fisiologia , Sistema de Sinalização das MAP Quinases , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta1/fisiologia , Regulação para Cima , Animais , Transdiferenciação Celular , Células Cultivadas , Expressão Gênica , Vasos Linfáticos/citologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas ras/metabolismo
12.
Biol Reprod ; 94(4): 92, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26984996

RESUMO

The F-box and leucine-rich repeat protein 10 (Fbxl10) gene encodes a protein that catalyzes demethylation of H3K4 and H3K36. In this study, we show the important roles of FBXL10 as a histone demethylase in sustainable sperm production using mice in which the JmjC domain of Fbxl10 was deleted (Fbxl10(DeltaJ/DeltaJ)). In histological analysis, testis sections from 10-wk-old Fbxl10(DeltaJ/DeltaJ) mice appeared normal. On the other hand, testes from 7-mo-old Fbxl10(DeltaJ/DeltaJ) mice contained a greater ratio of seminiferous tubules exhibiting degeneration of spermatogenesis. Further analysis using an in vitro spermatogonia culture system, that is, germline stem cells (GSCs), revealed that Fbxl10(DeltaJ/DeltaJ) GSCs expressed a significantly higher level of P21 and P19 mRNA, cyclin-dependent kinase inhibitors and also known as cellular senescence markers, than wild-type (WT) GSCs. Furthermore, the ratio of Fbxl10(DeltaJ/DeltaJ) GSCs in G0/G1 phase was higher and the ratios in S and G2/M phases were lower than the corresponding ratios of WT GSCs, and the doubling speed of Fbxl10(DeltaJ/DeltaJ) GSCs was significantly slower than that of WT GSCs. In addition to these in vitro results, an in vivo study indicated that recovery of spermatogenesis after a transient reduction in the number of testicular germ cells by busulfan treatment was significantly slower in Fbxl10(DeltaJ/DeltaJ) mice than in WT mice. These data suggest that Fbxl10 plays important roles in long-term sustainable spermatogenesis via regulating cell cycle.


Assuntos
Células-Tronco Germinativas Adultas/metabolismo , Proteínas F-Box/metabolismo , Histona Desmetilases com o Domínio Jumonji/metabolismo , Espermatogênese , Espermatogônias/fisiologia , Animais , Bussulfano , Ciclo Celular , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Testículo/metabolismo
13.
Exp Eye Res ; 152: 34-42, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27639517

RESUMO

5'TG3'-interacting factors (TGIFs) function as transcriptional repressors. Defects in TGIFs cause severe abnormalities in the developing brain and face. We found that Tgif2 was highly expressed in the mouse retina at early stages of development and examined its role in retinal development. Knockdown of Tgif2 in retinal explants at E14 using shRNA (sh-Tgif2) resulted in a decreased number of rod photoreceptors, whereas the number of cone photoreceptors increased without perturbation of cell proliferation and apoptosis. Concomitantly, the expression levels of photoreceptor-related genes were decreased in sh-Tgif2-introduced retinal explants. To examine the in vivo effects of Tgif2 overexpression, we generated Tgif2 conditional knock-in mice (Tgif2cKI). Although retinal cell differentiation, based on the relative proportions of retinal subtypes in the mature retina, was not affected, we observed abnormal localization of cone photoreceptor cell nuclei in the outer nuclear layer of the Tgif2cKI retina. However, electrical retinography suggest that cones in Tgif2cKI were functionally equivalent to those of wild mice. Our study revealed that Tgif2 participates in photoreceptor cell differentiation in the early stages of retinal development and regulates proper subretinal localization of the cone photoreceptors.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Células Fotorreceptoras de Vertebrados/metabolismo , RNA/genética , Proteínas Repressoras/genética , Retina/metabolismo , Animais , Modelos Animais de Doenças , Eletrorretinografia , Técnicas de Silenciamento de Genes , Proteínas de Homeodomínio/biossíntese , Hibridização In Situ , Camundongos , Camundongos Transgênicos , Células Fotorreceptoras de Vertebrados/patologia , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Repressoras/biossíntese , Doenças Retinianas/genética , Doenças Retinianas/metabolismo , Doenças Retinianas/patologia , Técnicas de Cultura de Tecidos
14.
Proc Natl Acad Sci U S A ; 110(51): 20699-704, 2013 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-24302768

RESUMO

High-mobility group box 1 (HMGB1) is a DNA-binding protein abundantly expressed in the nucleus that has gained much attention for its regulation of immunity and inflammation. Despite this, whether and how HMGB1 contributes to protective and/or pathological responses in vivo is unclear. In this study, we constructed Hmgb1-floxed (Hmgb1(f)(/f)) mice to achieve the conditional inactivation of the gene in a cell- and tissue-specific manner by crossing these mice with an appropriate Cre recombinase transgenic strain. Interestingly, although mice with HMGB1 ablation in myeloid cells apparently develop normally, they are more sensitive to endotoxin shock compared with control mice, which is accompanied by massive macrophage cell death. Furthermore, these mice also show an increased sensitivity to Listeria monocytogenes infection. We also provide evidence that the loss of HMGB1 in macrophages results in the suppression of autophagy, which is commonly induced by lipopolysaccharide stimulation or L. monocytogenes infection. Thus, intracellular HMGB1 contributes to the protection of mice from endotoxemia and bacterial infection by mediating autophagy in macrophages. These newly generated HMGB1 conditional knockout mice will serve a useful tool with which to study further the in vivo role of this protein in various pathological conditions.


Assuntos
Endotoxemia/imunologia , Proteína HMGB1/imunologia , Imunidade Inata , Listeria monocytogenes/imunologia , Listeriose/imunologia , Macrófagos/imunologia , Animais , Autofagia/genética , Autofagia/imunologia , Linhagem Celular , Endotoxemia/genética , Endotoxemia/metabolismo , Endotoxemia/patologia , Deleção de Genes , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Listeriose/genética , Listeriose/metabolismo , Listeriose/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Transgênicos
15.
EMBO J ; 30(22): 4678-91, 2011 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-21897366

RESUMO

Obesity is a life-threatening factor and is often associated with dysregulation of gene expression. Here, we show that the CNOT3 subunit of the CCR4-NOT deadenylase complex is critical to metabolic regulation. Cnot3(+/-) mice are lean with hepatic and adipose tissues containing reduced levels of lipids, and show increased metabolic rates and enhanced glucose tolerance. Cnot3(+/-) mice remain lean and sensitive to insulin even on a high-fat diet. Furthermore, introduction of Cnot3 haplodeficiency in ob/ob mice ameliorated the obese phenotype. Hepatic expression of most mRNAs is not altered in Cnot3(+/-) vis-à-vis wild-type mice. However, the levels of specific mRNAs, such as those coding for energy metabolism-related PDK4 and IGFBP1, are increased in Cnot3(+/-) hepatocytes, having poly(A) tails that are longer than those seen in control cells. We provide evidence that CNOT3 is involved in recruitment of the CCR4-NOT deadenylase to the 3' end of specific mRNAs. Finally, as CNOT3 levels in the liver and white adipose tissues decrease upon fasting, we propose that CNOT3 responds to feeding conditions to regulate deadenylation-specific mRNAs and energy metabolism.


Assuntos
Metabolismo Energético , Obesidade/genética , RNA Mensageiro/biossíntese , Fatores de Transcrição/metabolismo , Tecido Adiposo/metabolismo , Animais , Dieta , Insulina/metabolismo , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/biossíntese , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Fígado/metabolismo , Camundongos , Camundongos Obesos/genética , Camundongos Obesos/metabolismo , Camundongos Transgênicos , Dados de Sequência Molecular , Obesidade/metabolismo , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Serina-Treonina Quinases/genética , Piruvato Desidrogenase Quinase de Transferência de Acetil , RNA Mensageiro/genética , Fatores de Transcrição/genética
16.
Eur J Immunol ; 44(6): 1622-32, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24536025

RESUMO

The intracellular adaptor Lnk (also known as SH2B3) regulates cytokine signals that control lymphohematopoiesis, and Lnk(-/-) mice have expanded B-cell, megakaryocyte, and hematopoietic stem-cell populations. Moreover, mutations in the LNK gene are found in patients with myeloproliferative disease, whereas LNK polymorphisms have recently been associated with inflammatory and autoimmune diseases, including celiac disease. Here, we describe a previously unrecognized function of Lnk in the control of inflammatory CD8(+) T-cell proliferation and in intestinal homeostasis. Mature T cells from newly generated Lnk-Venus reporter mice had low but substantial expression of Lnk, whereas Lnk expression was downregulated during homeostatic T-cell proliferation under lymphopenic conditions. The numbers of CD44(hi) IFN-γ(+) CD8(+) effector or memory T cells were found to be increased in Lnk(-/-) mice, which also exhibited shortening of villi in the small intestine. Lnk(-/-) CD8(+) T cells survived longer in response to stimulation with IL-15 and proliferated even in nonlymphopenic hosts. Transfer of Lnk(-/-) CD8(+) T cells together with WT CD4(+) T cells into Rag2-deficient mice recapitulated a sign of villous abnormality. Our results reveal a link between Lnk and immune cell-mediated intestinal tissue destruction.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Homeostase/imunologia , Intestinos/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Linfócitos T CD8-Positivos/patologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Homeostase/genética , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/imunologia , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Intestinos/patologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana , Camundongos , Camundongos Knockout , Polimorfismo Genético/imunologia
17.
Genesis ; 52(5): 440-50, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24700560

RESUMO

The introduction of a transgene into the genome through homologous recombination or sequence-specific enzymatic modification is a key technique for producing transgenic mice. The Rosa26 gene has been widely used to produce transgenic mice because the gene is transcriptionally active in various cell types and, at many developmental stages, is permissive for constitutive expression of integrated transgenes, and is dispensable for normal development. However, permissive loci other than Rosa26 are needed to generate mice that harbor multiple transgenes for complex studies. Here, we identified the Cd6 locus on mouse chromosome 19 as a transgene integration site in a transgenic mouse strain showing widespread reporter expression. Using this locus, we generated a knock-in mouse line that harbors a CAG promoter-driven reporter transgene, and found that the homozygous transgenic mice are viable and fertile, although transgene insertion disrupted Cd6 gene expression. The transgene on the Cd6 locus expressed reporter genes extensively throughout embryos, neonates, and adults. Combined with the Cre/loxP binary system, blood and lymphatic endothelial cell-specific reporter expression from the transgenic locus was achieved. These results suggest that Cd6 is valuable as an alternative site for targeted transgenesis.


Assuntos
Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Diferenciação de Linfócitos T/genética , Antígenos de Diferenciação de Linfócitos T/metabolismo , Transgenes , Animais , Regulação da Expressão Gênica , Técnicas de Introdução de Genes , Genes Reporter , Loci Gênicos , Íntrons , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Recombinação Genética
18.
J Biol Chem ; 288(25): 18546-60, 2013 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-23649632

RESUMO

Human embryonic stem cells and mouse epiblast stem cells represent a primed pluripotent stem cell state that requires TGF-ß/activin signaling. TGF-ß and/or activin are commonly thought to regulate transcription through both Smad2 and Smad3. However, the different contributions of these two Smads to primed pluripotency and the downstream events that they may regulate remain poorly understood. We addressed the individual roles of Smad2 and Smad3 in the maintenance of primed pluripotency. We found that Smad2, but not Smad3, is required to maintain the undifferentiated pluripotent state. We defined a Smad2 regulatory circuit in human embryonic stem cells and mouse epiblast stem cells, in which Smad2 acts through binding to regulatory promoter sequences to activate Nanog expression while in parallel repressing autocrine bone morphogenetic protein signaling. Increased autocrine bone morphogenetic protein signaling caused by Smad2 down-regulation leads to cell differentiation toward the trophectoderm, mesoderm, and germ cell lineages. Additionally, induction of Cdx2 expression, as a result of decreased Smad2 expression, leads to repression of Oct4 expression, which, together with the decreased Nanog expression, accelerates the loss of pluripotency. These findings reveal that Smad2 is a unique integrator of transcription and signaling events and is essential for the maintenance of the mouse and human primed pluripotent stem cell state.


Assuntos
Células-Tronco Embrionárias/metabolismo , Camadas Germinativas/metabolismo , Células-Tronco Pluripotentes/metabolismo , Proteína Smad2/metabolismo , Animais , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Fator de Transcrição CDX2 , Diferenciação Celular/genética , Células Cultivadas , Células-Tronco Embrionárias/citologia , Camadas Germinativas/citologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Immunoblotting , Camundongos , Camundongos da Linhagem 129 , Microscopia de Fluorescência , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco Pluripotentes/citologia , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Proteína Smad2/genética , Proteína Smad3/genética , Proteína Smad3/metabolismo , Transcrição Gênica , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
19.
Development ; 138(23): 5235-46, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22069191

RESUMO

Mastermind (Mam) is one of the elements of Notch signaling, a system that plays a pivotal role in metazoan development. Mam proteins form transcriptionally activating complexes with the intracellular domains of Notch, which are generated in response to the ligand-receptor interaction, and CSL DNA-binding proteins. In mammals, three structurally divergent Mam isoforms (MamL1, MamL2 and MamL3) have been identified. There have also been indications that Mam interacts functionally with various other transcription factors, including the p53 tumor suppressor, ß-catenin and NF-κB. We have demonstrated previously that disruption of MamL1 causes partial deficiency of Notch signaling in vivo. However, MamL1-deficient mice did not recapitulate total loss of Notch signaling, suggesting that other members could compensate for the loss or that Notch signaling could proceed in the absence of Mam in certain contexts. Here, we report the generation of lines of mice null for MamL3. Although MamL3-null mice showed no apparent abnormalities, mice null for both MamL1 and MamL3 died during the early organogenic period with classic pan-Notch defects. Furthermore, expression of the lunatic fringe gene, which is strictly controlled by Notch signaling in the posterior presomitic mesoderm, was undetectable in this tissue of the double-null embryos. Neither of the single-null embryos exhibited any of these phenotypes. These various roles of the three Mam proteins could be due to their differential physical characteristics and/or their spatiotemporal distributions. These results indicate that engagement of Mam is essential for Notch signaling, and that the three Mam isoforms have distinct roles in vivo.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas Nucleares/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Animais , Southern Blotting , Western Blotting , Primers do DNA/genética , Fibroblastos , Citometria de Fluxo , Regulação da Expressão Gênica no Desenvolvimento/genética , Glicosiltransferases/metabolismo , Hibridização In Situ , Luciferases , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteínas Nucleares/genética , Plasmídeos/genética , Reação em Cadeia da Polimerase , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/genética , Transativadores/genética , Fatores de Transcrição/genética
20.
Cereb Cortex ; 23(8): 1824-35, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22705452

RESUMO

Polypyrimidine tract-binding protein (PTB) is a well-characterized RNA-binding protein and known to be preferentially expressed in neural stem cells (NSCs) in the central nervous system; however, its role in NSCs in the developing brain remains unclear. To explore the role of PTB in embryonic NSCs in vivo, Nestin-Cre-mediated conditional Ptb knockout mice were generated for this study. In the mutant forebrain, despite the depletion of PTB protein, neither abnormal neurogenesis nor flagrant morphological abnormalities were observed at embryonic day 14.5 (E14.5). Nevertheless, by 10 weeks, nearly all mutant mice succumbed to hydrocephalus (HC), which was caused by a lack of the ependymal cell layer in the dorsal cortex. Upon further analysis, a gradual loss of adherens junctions (AJs) was observed in the ventricular zone (VZ) of the dorsal telencephalon in the mutant brains, beginning at E14.5. In the AJs-deficient VZ, impaired interkinetic nuclear migration and precocious differentiation of NSCs were observed after E14.5. These findings demonstrated that PTB depletion in the dorsal telencephalon is causally involved in the development of HC and that PTB is important for the maintenance of AJs in the NSCs of the dorsal telencephalon.


Assuntos
Junções Aderentes/ultraestrutura , Hidrocefalia/etiologia , Proteína de Ligação a Regiões Ricas em Polipirimidinas/fisiologia , Telencéfalo/embriologia , Animais , Hidrocefalia/genética , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Neurais/ultraestrutura , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Telencéfalo/anormalidades
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA