Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Development ; 148(10)2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33998651

RESUMEN

Heterochromatin-related epigenetic mechanisms, such as DNA methylation, facilitate pairing of homologous chromosomes during the meiotic prophase of mammalian spermatogenesis. In pro-spermatogonia, de novo DNA methylation plays a key role in completing meiotic prophase and initiating meiotic division. However, the role of maintenance DNA methylation in the regulation of meiosis, especially in the adult, is not well understood. Here, we reveal that NP95 (also known as UHRF1) and DNMT1 - two essential proteins for maintenance DNA methylation - are co-expressed in spermatogonia and are necessary for meiosis in male germ cells. We find that Np95- or Dnmt1-deficient spermatocytes exhibit spermatogenic defects characterized by synaptic failure during meiotic prophase. In addition, assembly of pericentric heterochromatin clusters in early meiotic prophase, a phenomenon that is required for subsequent pairing of homologous chromosomes, is disrupted in both mutants. Based on these observations, we propose that DNA methylation, established in pre-meiotic spermatogonia, regulates synapsis of homologous chromosomes and, in turn, quality control of male germ cells. Maintenance DNA methylation, therefore, plays a role in ensuring faithful transmission of both genetic and epigenetic information to offspring.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Emparejamiento Cromosómico/genética , ADN (Citosina-5-)-Metiltransferasa 1/genética , Metilación de ADN/genética , Espermatocitos/crecimiento & desarrollo , Espermatogénesis/genética , Ubiquitina-Proteína Ligasas/genética , Células Madre Germinales Adultas/citología , Animales , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Epigénesis Genética/genética , Heterocromatina/metabolismo , Masculino , Ratones , Ratones Noqueados , Espermatocitos/fisiología , Espermatogénesis/fisiología , Ubiquitina-Proteína Ligasas/metabolismo
2.
Dis Model Mech ; 12(11)2019 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-31562139

RESUMEN

Respiratory failure is a life-threatening problem for pre-term and term infants, yet many causes remain unknown. Here, we present evidence that whey acidic protein (WAP) four-disulfide core domain protease inhibitor 2 (Wfdc2), a protease inhibitor previously unrecognized in respiratory disease, may be a causal factor in infant respiratory failure. Wfdc2 transcripts are detected in the embryonic lung and analysis of a Wfdc2-GFP knock-in mouse line shows that both basal and club cells, and type II alveolar epithelial cells (AECIIs), express Wfdc2 neonatally. Wfdc2-null-mutant mice display progressive atelectasis after birth with a lethal phenotype. Mutant lungs have multiple defects, including impaired cilia and the absence of mature club cells from the tracheo-bronchial airways, and malformed lamellar bodies in AECIIs. RNA sequencing shows significant activation of a pro-inflammatory pathway, but with low-quantity infiltration of mononuclear cells in the lung. These data demonstrate that Wfdc2 function is vitally important for lung aeration at birth and that gene deficiency likely causes failure of the lung mucosal barrier.


Asunto(s)
Insuficiencia Respiratoria/mortalidad , Proteína 2 de Dominio del Núcleo de Cuatro Disulfuros WAP/fisiología , Animales , Animales Recién Nacidos , Diferenciación Celular , Células Cultivadas , Cilios/fisiología , Humanos , Ratones , Ratones Endogámicos ICR , Atelectasia Pulmonar/etiología , Surfactantes Pulmonares/metabolismo
3.
Development ; 145(23)2018 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-30504434

RESUMEN

The mammalian male germline is sustained by a pool of spermatogonial stem cells (SSCs) that can transmit both genetic and epigenetic information to offspring. However, the mechanisms underlying epigenetic transmission remain unclear. The histone methyltransferase Kmt2b is highly expressed in SSCs and is required for the SSC-to-progenitor transition. At the stem-cell stage, Kmt2b catalyzes H3K4me3 at bivalent H3K27me3-marked promoters as well as at promoters of a new class of genes lacking H3K27me3, which we call monovalent. Monovalent genes are mainly activated in late spermatogenesis, whereas most bivalent genes are mainly not expressed until embryonic development. These data suggest that SSCs are epigenetically primed by Kmt2b in two distinguishable ways for the upregulation of gene expression both during the spermatogenic program and through the male germline into the embryo. Because Kmt2b is also the major H3K4 methyltransferase for bivalent promoters in embryonic stem cells, we also propose that Kmt2b has the capacity to prime stem cells epigenetically.


Asunto(s)
Embrión de Mamíferos/metabolismo , Células Germinativas/citología , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Regiones Promotoras Genéticas , Espermatogonias/citología , Células Madre/citología , Células Madre/metabolismo , Animales , Supervivencia Celular , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , Masculino , Ratones , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas del Grupo Polycomb/metabolismo
4.
Front Cell Neurosci ; 11: 143, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28567004

RESUMEN

Potential clinical applications of neurons derived from human induced pluripotent stem cells (hiPSC-neurons) for drug screening and transplantation therapies have received considerable attention. However, it remains unclear whether and how transplanted hiPSC-neurons are incorporated into pre-existing neural circuits. Here we developed a co-culture system of hiPSC-neurons and mouse hippocampal slices to examine the differentiation of hiPSC-neurons in pre-existing neural circuits. hiPSC-neurons transplanted in mouse hippocampal slices expressed the hippocampal neuron-specific markers HuB and Prox1 after 7 days of culture, while those markers were scarcely expressed in hiPSC-neurons cultured on glass dishes. Furthermore, hiPSC-neurons transplanted in the dentate gyrus (DG) of slice cultures grew to exhibit dentate granule cell-like morphologies, including besom-shaped dendrites. Similarly, hiPSC-neurons transplanted in the CA1 region of slice cultures grew to exhibit CA1 pyramidal cell-like morphologies, including primary apical and multiple basal dendrites with synaptic spines. Additionally, these cells projected axons toward the entorhinal cortex (EC) as observed in vivo. These data suggest that hiPSC-neurons were anatomically integrated into pre-existing neural circuits in a region-specific manner. Thus, the co-culture system will be useful for the study of efficient strategies to differentiate transplanted hiPSC-neurons.

5.
PLoS One ; 12(2): e0171947, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28192470

RESUMEN

Oligodendrocytes myelinate axons and form myelin sheaths in the central nervous system. The development of therapies for demyelinating diseases, including multiple sclerosis and leukodystrophies, is a challenge because the pathogenic mechanisms of disease remain poorly understood. Primate pluripotent stem cell-derived oligodendrocytes are expected to help elucidate the molecular pathogenesis of these diseases. Oligodendrocytes have been successfully differentiated from human pluripotent stem cells. However, it is challenging to prepare large amounts of oligodendrocytes over a short amount of time because of manipulation difficulties under conventional primate pluripotent stem cell culture methods. We developed a proprietary dissociated monolayer and feeder-free culture system to handle pluripotent stem cell cultures. Because the dissociated monolayer and feeder-free culture system improves the quality and growth of primate pluripotent stem cells, these cells could potentially be differentiated into any desired functional cells and consistently cultured in large-scale conditions. In the current study, oligodendrocyte progenitor cells and mature oligodendrocytes were generated within three months from monkey embryonic stem cells. The embryonic stem cell-derived oligodendrocytes exhibited in vitro myelinogenic potency with rat dorsal root ganglion neurons. Additionally, the transplanted oligodendrocyte progenitor cells differentiated into myelin basic protein-positive mature oligodendrocytes in the mouse corpus callosum. This preparative method was used for human induced pluripotent stem cells, which were also successfully differentiated into oligodendrocyte progenitor cells and mature oligodendrocytes that were capable of myelinating rat dorsal root ganglion neurons. Moreover, it was possible to freeze, thaw, and successfully re-culture the differentiating cells. These results showed that embryonic stem cells and human induced pluripotent stem cells maintained in a dissociated monolayer and feeder-free culture system have the potential to generate oligodendrocyte progenitor cells and mature oligodendrocytes in vitro and in vivo. This culture method could be applied to prepare large amounts of oligodendrocyte progenitor cells and mature oligodendrocytes in a relatively short amount of time.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Oligodendroglía/citología , Células Madre Pluripotentes/citología , Células Madre/citología , Animales , Western Blotting , Células Cultivadas , Técnicas de Cocultivo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Células Nutrientes , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Macaca fascicularis , Ratones Endogámicos C57BL , Microscopía Fluorescente , Proteína Básica de Mielina/metabolismo , Vaina de Mielina/metabolismo , Neuronas/citología , Neuronas/metabolismo , Oligodendroglía/metabolismo , Células Madre Pluripotentes/metabolismo , Ratas Sprague-Dawley , Trasplante de Células Madre/métodos , Células Madre/metabolismo
6.
BMC Genomics ; 16: 624, 2015 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-26290333

RESUMEN

BACKGROUND: In the male germline, neonatal prospermatogonia give rise to spermatogonia, which include stem cell population (undifferentiated spermatogonia) that supports continuous spermatogenesis in adults. Although the levels of DNA methyltransferases change dynamically in the neonatal and early postnatal male germ cells, detailed genome-wide DNA methylation profiles of these cells during the stem cell formation and differentiation have not been reported. RESULTS: To understand the regulation of spermatogonial stem cell formation and differentiation, we examined the DNA methylation and gene expression dynamics of male mouse germ cells at the critical stages: neonatal prospermatogonia, and early postntal (day 7) undifferentiated and differentiating spermatogonia. We found large partially methylated domains similar to those found in cancer cells and placenta in all these germ cells, and high levels of non-CG methylation and 5-hydroxymethylcytosines in neonatal prospermatogonia. Although the global CG methylation levels were stable in early postnatal male germ cells, and despite the reported scarcity of differential methylation in the adult spermatogonial stem cells, we identified many regions showing stage-specific differential methylation in and around genes important for stem cell function and spermatogenesis. These regions contained binding sites for specific transcription factors including the SOX family members. CONCLUSIONS: Our findings show a distinctive and dynamic regulation of DNA methylation during spermatogonial stem cell formation and differentiation in the neonatal and early postnatal testes. Furthermore, we revealed a unique accumulation and distribution of non-CG methylation and 5hmC marks in neonatal prospermatogonia. These findings contrast with the reported scarcity of differential methylation in adult spermatogonial stem cell differentiation and represent a unique phase of male germ cell development.


Asunto(s)
Metilación de ADN , Perfilación de la Expresión Génica/métodos , Espermatogonias/citología , Células Madre/fisiología , Animales , Animales Recién Nacidos , Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Espermatogénesis , Espermatogonias/fisiología
7.
Dev Cell ; 34(1): 96-107, 2015 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-26120033

RESUMEN

In many adult tissues, homeostasis relies on self-renewing stem cells that are primed for differentiation. The reconciliation mechanisms of these characteristics remain a fundamental question in stem cell biology. We propose that regulation at the post-transcriptional level is essential for homeostasis in murine spermatogonial stem cells (SSCs). Here, we show that Nanos2, an evolutionarily conserved RNA-binding protein, works with other cellular messenger ribonucleoprotein (mRNP) components to ensure the primitive status of SSCs through a dual mechanism that involves (1) direct recruitment and translational repression of genes that promote spermatogonial differentiation and (2) repression of the target of rapamycin complex 1 (mTORC1), a well-known negative pathway for SSC self-renewal, by sequestration of the core factor mTOR in mRNPs. This mechanism links mRNA turnover to mTORC1 signaling through Nanos2-containing mRNPs and establishes a post-transcriptional buffering system to facilitate SSC homeostasis in the fluctuating environment within the seminiferous tubule.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Proteínas de Unión al ARN/genética , ARN/metabolismo , Espermatogonias/citología , Células Madre/citología , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Masculino , Ratones , Transducción de Señal/fisiología
8.
J Pharmacol Sci ; 126(3): 230-42, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25341845

RESUMEN

Intracranial aneurysm (IA) and aortic dissection are both complications of hypertension and characterized by degeneration of the media. Given the involvement of prostaglandin F2α and its receptor, FP, in extracellular matrix remodeling in a mouse model of pulmonary fibrosis, here we induced hypertension and IA in rats by salt loading and hemi-lateral ligation of renal and carotid arteries and examined effects of a selective FP antagonist, AS604872, on these vascular events. AS604872 significantly accelerated degeneration of the media in both cerebral artery and aorta as evidenced by thinning of the media and disruption of the elastic lamina and promoted IA and aortic dissection. Notably, AS604872 induced expression of pro-inflammatory genes such as E-selectin in lesions and significantly enhanced macrophage infiltration. Suppression of surface expression of E-selectin with cimetidine prevented macrophage infiltration and aortic dissection. Thus, AS604872 exacerbates vascular inflammation in hypertensive rats and facilitates IA and aortic dissection. These results demonstrate that both IA and aortic dissection are caused by chronic inflammation of the arterial wall, which is worsened by AS604872, cautioning that other FP antagonists may share such deleterious actions in vascular homeostasis and suggesting that AS604872 can be used to make models of these vascular diseases with extensive degeneration.


Asunto(s)
Aneurisma de la Aorta/inducido químicamente , Disección Aórtica/inducido químicamente , Compuestos de Bifenilo/toxicidad , Hipertensión/complicaciones , Aneurisma Intracraneal/inducido químicamente , Antagonistas de Prostaglandina/toxicidad , Receptores de Prostaglandina/antagonistas & inhibidores , Sulfonamidas/toxicidad , Disección Aórtica/genética , Disección Aórtica/metabolismo , Disección Aórtica/patología , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Aorta/patología , Aneurisma de la Aorta/genética , Aneurisma de la Aorta/metabolismo , Aneurisma de la Aorta/patología , Arterias Cerebrales/efectos de los fármacos , Arterias Cerebrales/metabolismo , Arterias Cerebrales/patología , Modelos Animales de Enfermedad , Colágenos Fibrilares/metabolismo , Humanos , Hipertensión/genética , Hipertensión/metabolismo , Mediadores de Inflamación/metabolismo , Aneurisma Intracraneal/genética , Aneurisma Intracraneal/metabolismo , Aneurisma Intracraneal/patología , Masculino , Ratones Noqueados , Ratas Sprague-Dawley , Receptores de Prostaglandina/genética , Receptores de Prostaglandina/metabolismo , Cloruro de Sodio Dietético , Factores de Tiempo , Remodelación Vascular/efectos de los fármacos , Vasculitis/inducido químicamente , Vasculitis/metabolismo , Vasculitis/patología
9.
Development ; 140(17): 3565-76, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23903187

RESUMEN

Epigenetic modifications influence gene expression and chromatin remodeling. In embryonic pluripotent stem cells, these epigenetic modifications have been extensively characterized; by contrast, the epigenetic events of tissue-specific stem cells are poorly understood. Here, we define a new epigenetic shift that is crucial for differentiation of murine spermatogonia toward meiosis. We have exploited a property of incomplete cytokinesis, which causes male germ cells to form aligned chains of characteristic lengths, as they divide and differentiate. These chains revealed the stage of spermatogenesis, so the epigenetic differences of various stages could be characterized. Single, paired and medium chain-length spermatogonia not expressing Kit (a marker of differentiating spermatogonia) showed no expression of Dnmt3a2 and Dnmt3b (two de novo DNA methyltransferases); they also lacked the transcriptionally repressive histone modification H3K9me2. By contrast, spermatogonia consisting of ~8-16 chained cells with Kit expression dramatically upregulated Dnmt3a2/3b expression and also displayed increased H3K9me2 modification. To explore the function of these epigenetic changes in spermatogonia in vivo, the DNA methylation machinery was destabilized by ectopic Dnmt3b expression or Np95 ablation. Forced Dnmt3b expression induced expression of Kit; whereas ablation of Np95, which is essential for maintaining DNA methylation, interfered with differentiation and viability only after spermatogonia become Kit positive. These data suggest that the epigenetic status of spermatogonia shifts dramatically during the Kit-negative to Kit-positive transition. This shift might serve as a switch that determines whether spermatogonia self-renew or differentiate.


Asunto(s)
Diferenciación Celular/fisiología , Epigénesis Genética/fisiología , Células Germinativas/fisiología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Espermatogénesis/fisiología , Espermatogonias/crecimiento & desarrollo , Animales , Western Blotting , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/fisiología , Cartilla de ADN/genética , Citometría de Flujo , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Espermatogonias/citología , ADN Metiltransferasa 3B
10.
Development ; 138(19): 4207-17, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21896631

RESUMEN

During meiosis, specific histone modifications at pericentric heterochromatin (PCH), especially histone H3 tri- and dimethylation at lysine 9 (H3K9me3 and H3K9me2, respectively), are required for proper chromosome interactions. However, the molecular mechanism by which H3K9 methylation mediates the synapsis is not yet understood. We have generated a Cbx3-deficient mouse line and performed comparative analysis on Suv39h1/h2-, G9a- and Cbx3-deficient spermatocytes. This study revealed that H3K9me2 at PCH depended on Suv39h1/h2-mediated H3K9me3 and its recognition by the Cbx3 gene product HP1γ. We further found that centromere clustering and synapsis were commonly affected in G9a- and Cbx3-deficient spermatocytes. These genetic observations suggest that HP1γ/G9a-dependent PCH-mediated centromere clustering is an axis for proper chromosome interactions during meiotic prophase. We propose that the role of the HP1γ/G9a axis is to retain centromeric regions of unpaired homologous chromosomes in close alignment and facilitate progression of their pairing in early meiotic prophase. This study also reveals considerable plasticity in the interplay between different histone modifications and suggests that such stepwise and dynamic epigenetic modifications may play a pivotal role in meiosis.


Asunto(s)
Proteínas Cromosómicas no Histona/genética , Emparejamiento Cromosómico , N-Metiltransferasa de Histona-Lisina/genética , Histonas/química , Meiosis , Animales , Centrómero/ultraestructura , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/fisiología , Epigénesis Genética , Femenino , N-Metiltransferasa de Histona-Lisina/fisiología , Masculino , Metilación , Ratones , Ratones Noqueados , Microscopía Fluorescente/métodos , Modelos Genéticos , Espermatocitos/citología
11.
PLoS Comput Biol ; 5(6): e1000397, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19503826

RESUMEN

Predictions of interactions between target proteins and potential leads are of great benefit in the drug discovery process. We present a comprehensively applicable statistical prediction method for interactions between any proteins and chemical compounds, which requires only protein sequence data and chemical structure data and utilizes the statistical learning method of support vector machines. In order to realize reasonable comprehensive predictions which can involve many false positives, we propose two approaches for reduction of false positives: (i) efficient use of multiple statistical prediction models in the framework of two-layer SVM and (ii) reasonable design of the negative data to construct statistical prediction models. In two-layer SVM, outputs produced by the first-layer SVM models, which are constructed with different negative samples and reflect different aspects of classifications, are utilized as inputs to the second-layer SVM. In order to design negative data which produce fewer false positive predictions, we iteratively construct SVM models or classification boundaries from positive and tentative negative samples and select additional negative sample candidates according to pre-determined rules. Moreover, in order to fully utilize the advantages of statistical learning methods, we propose a strategy to effectively feedback experimental results to computational predictions with consideration of biological effects of interest. We show the usefulness of our approach in predicting potential ligands binding to human androgen receptors from more than 19 million chemical compounds and verifying these predictions by in vitro binding. Moreover, we utilize this experimental validation as feedback to enhance subsequent computational predictions, and experimentally validate these predictions again. This efficient procedure of the iteration of the in silico prediction and in vitro or in vivo experimental verifications with the sufficient feedback enabled us to identify novel ligand candidates which were distant from known ligands in the chemical space.


Asunto(s)
Inteligencia Artificial , Simulación por Computador , Descubrimiento de Drogas/métodos , Modelos Estadísticos , Proteínas , Algoritmos , Secuencia de Aminoácidos , Área Bajo la Curva , Sitios de Unión , Bases de Datos de Proteínas , Retroalimentación , Ligandos , Modelos Químicos , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Proteínas/química , Proteínas/metabolismo , Receptores Androgénicos/química , Receptores Androgénicos/metabolismo , Reproducibilidad de los Resultados , Análisis de Secuencia de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA