Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 242
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 615(7954): 900-906, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36922585

RESUMEN

Sex chromosome disorders severely compromise gametogenesis in both males and females. In oogenesis, the presence of an additional Y chromosome or the loss of an X chromosome disturbs the robust production of oocytes1-5. Here we efficiently converted the XY chromosome set to XX without an additional Y chromosome in mouse pluripotent stem (PS) cells. In addition, this chromosomal alteration successfully eradicated trisomy 16, a model of Down's syndrome, in PS cells. Artificially produced euploid XX PS cells differentiated into mature oocytes in culture with similar efficiency to native XX PS cells. Using this method, we differentiated induced pluripotent stem cells from the tail of a sexually mature male mouse into fully potent oocytes, which gave rise to offspring after fertilization. This study provides insights that could ameliorate infertility caused by sex chromosome or autosomal disorders, and opens the possibility of bipaternal reproduction.


Asunto(s)
Ingeniería Genética , Técnicas In Vitro , Oocitos , Cromosoma X , Animales , Femenino , Masculino , Ratones , Oocitos/metabolismo , Oocitos/fisiología , Cromosoma X/genética , Cromosoma Y/genética , Células Madre Pluripotentes/metabolismo , Síndrome de Down/genética , Síndrome de Down/terapia , Fertilización , Infertilidad/terapia , Homosexualidad Masculina , Trastornos de los Cromosomas Sexuales/complicaciones , Trastornos de los Cromosomas Sexuales/genética , Trastornos de los Cromosomas Sexuales/terapia , Ingeniería Genética/métodos
2.
EMBO J ; 41(18): e110815, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35912849

RESUMEN

In vitro oogenesis is key to elucidating the mechanism of human female germ-cell development and its anomalies. Accordingly, pluripotent stem cells have been induced into primordial germ cell-like cells and into oogonia with epigenetic reprogramming, yet further reconstitutions remain a challenge. Here, we demonstrate ex vivo reconstitution of fetal oocyte development in both humans and cynomolgus monkeys (Macaca fascicularis). With an optimized culture of fetal ovary reaggregates over three months, human and monkey oogonia enter and complete the first meiotic prophase to differentiate into diplotene oocytes that form primordial follicles, the source for oogenesis in adults. The cytological and transcriptomic progressions of fetal oocyte development in vitro closely recapitulate those in vivo. A comparison of single-cell transcriptomes among humans, monkeys, and mice unravels primate-specific and conserved programs driving fetal oocyte development, the former including a distinct transcriptomic transformation upon oogonia-to-oocyte transition and the latter including two active X chromosomes with little X-chromosome upregulation. Our study provides a critical step forward for realizing human in vitro oogenesis and uncovers salient characteristics of fetal oocyte development in primates.


Asunto(s)
Meiosis , Oogénesis , Animales , Femenino , Humanos , Macaca fascicularis , Ratones , Oocitos , Oogénesis/fisiología , Ovario
3.
EMBO J ; 41(13): e110600, 2022 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-35703121

RESUMEN

Germ cells are unique in engendering totipotency, yet the mechanisms underlying this capacity remain elusive. Here, we perform comprehensive and in-depth nucleome analysis of mouse germ-cell development in vitro, encompassing pluripotent precursors, primordial germ cells (PGCs) before and after epigenetic reprogramming, and spermatogonia/spermatogonial stem cells (SSCs). Although epigenetic reprogramming, including genome-wide DNA de-methylation, creates broadly open chromatin with abundant enhancer-like signatures, the augmented chromatin insulation safeguards transcriptional fidelity. These insulatory constraints are then erased en masse for spermatogonial development. Notably, despite distinguishing epigenetic programming, including global DNA re-methylation, the PGCs-to-spermatogonia/SSCs development entails further euchromatization. This accompanies substantial erasure of lamina-associated domains, generating spermatogonia/SSCs with a minimal peripheral attachment of chromatin except for pericentromeres-an architecture conserved in primates. Accordingly, faulty nucleome maturation, including persistent insulation and improper euchromatization, leads to impaired spermatogenic potential. Given that PGCs after epigenetic reprogramming serve as oogenic progenitors as well, our findings elucidate a principle for the nucleome programming that creates gametogenic progenitors in both sexes, defining a basis for nuclear totipotency.


Asunto(s)
Epigénesis Genética , Células Germinativas , Animales , Cromatina/genética , Cromatina/metabolismo , Metilación de ADN , Epigenómica , Femenino , Células Germinativas/metabolismo , Masculino , Mamíferos/genética , Ratones , Espermatogonias
4.
Cell ; 146(4): 519-32, 2011 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-21820164

RESUMEN

The generation of properly functioning gametes in vitro requires reconstitution of the multistepped pathway of germ cell development. We demonstrate here the generation of primordial germ cell-like cells (PGCLCs) in mice with robust capacity for spermatogenesis. PGCLCs were generated from embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) through epiblast-like cells (EpiLCs), a cellular state highly similar to pregastrulating epiblasts but distinct from epiblast stem cells (EpiSCs). Reflecting epiblast development, EpiLC induction from ESCs/iPSCs is a progressive process, and EpiLCs highly competent for the PGC fate are a transient entity. The global transcription profiles, epigenetic reprogramming, and cellular dynamics during PGCLC induction from EpiLCs meticulously capture those associated with PGC specification from the epiblasts. Furthermore, we identify Integrin-ß3 and SSEA1 as markers that allow the isolation of PGCLCs with spermatogenic capacity from tumorigenic undifferentiated cells. Our findings provide a paradigm for the first step of in vitro gametogenesis.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Animales , Técnicas de Cultivo de Célula , Proliferación Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Perfilación de la Expresión Génica , Células Germinativas/citología , Células Germinativas/metabolismo , Estratos Germinativos/citología , Estratos Germinativos/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Espermatogénesis
5.
EMBO J ; 39(21): e104929, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-32954504

RESUMEN

Human germ cells perpetuate human genetic and epigenetic information. However, the underlying mechanism remains elusive, due to a lack of appropriate experimental systems. Here, we show that human primordial germ cell-like cells (hPGCLCs) derived from human-induced pluripotent stem cells (hiPSCs) can be propagated to at least ~106 -fold over a period of 4 months under a defined condition in vitro. During expansion, hPGCLCs maintain an early hPGC-like transcriptome and preserve their genome-wide DNA methylation profiles, most likely due to retention of maintenance DNA methyltransferase activity. These characteristics contrast starkly with those of mouse PGCLCs, which, under an analogous condition, show a limited propagation (up to ~50-fold) and persist only around 1 week, yet undergo cell-autonomous genome-wide DNA demethylation. Importantly, upon aggregation culture with mouse embryonic ovarian somatic cells in xenogeneic-reconstituted ovaries, expanded hPGCLCs initiate genome-wide DNA demethylation and differentiate into oogonia/gonocyte-like cells, demonstrating their germline potential. By creating a paradigm for hPGCLC expansion, our study uncovers critical divergences in expansion potential and the mechanism for epigenetic reprogramming between the human and mouse germ cell lineage.


Asunto(s)
Células Germinativas/metabolismo , Ovario/embriología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Animales , Línea Celular , Desmetilación del ADN , Metilación de ADN , Células Madre Embrionarias/metabolismo , Epigénesis Genética , Epigenómica , Femenino , Genoma , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones
6.
Int Immunol ; 35(7): 313-326, 2023 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-36933193

RESUMEN

Using a zoobiquity concept, we directly connect animal phenotypes to a human disease mechanism: the reduction of local plasminogen levels caused by matrix metalloproteinase-9 (MMP9) activity is associated with the development of inflammation in the intestines of dogs and patients with inflammatory bowel disease. We first investigated inflammatory colorectal polyps (ICRPs), which are a canine gastrointestinal disease characterized by the presence of idiopathic chronic inflammation, in Miniature Dachshund (MD) and found 31 missense disease-associated SNPs by whole-exome sequencing. We sequenced them in 10 other dog breeds and found five, PLG, TCOF1, TG, COL9A2 and COL4A4, only in MD. We then investigated two rare and breed-specific missense SNPs (T/T SNPs), PLG: c.477G > T and c.478A>T, and found that ICRPs with the T/T SNP risk alleles showed less intact plasminogen and plasmin activity in the lesions compared to ICRPs without the risk alleles but no differences in serum. Moreover, we show that MMP9, which is an NF-κB target, caused the plasminogen reduction and that intestinal epithelial cells expressing plasminogen molecules were co-localized with epithelial cells expressing MMP9 in normal colons with the risk alleles. Importantly, MMP9 expression in patients with ulcerous colitis or Crohn's disease also co-localized with epithelial cells showing enhanced NF-κB activation and less plasminogen expression. Overall, our zoobiquity experiments showed that MMP9 induces the plasminogen reduction in the intestine, contributing to the development of local inflammation and suggesting the local MMP9-plasminogen axis is a therapeutic target in both dogs and patients. Therefore, zoobiquity-type experiments could bring new perspectives for biomarkers and therapeutic targets.


Asunto(s)
Enfermedades Inflamatorias del Intestino , Metaloproteinasa 9 de la Matriz , Humanos , Perros , Animales , Plasminógeno , FN-kappa B , Inflamación , Serina Proteasas
7.
Cell ; 137(3): 571-84, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19410550

RESUMEN

Specification of the germ cell lineage is vital to development and heredity. In mice, the germ cell fate is induced in pluripotent epiblast cells by signaling molecules, yet the underlying mechanism remains unknown. Here we demonstrate that germ cell fate in the epiblast is a direct consequence of Bmp4 signaling from the extraembryonic ectoderm (ExE), which is antagonized by the anterior visceral endoderm (AVE). Strikingly, Bmp8b from the ExE restricts AVE development, thereby contributing to Bmp4 signaling. Furthermore, Wnt3 in the epiblast ensures its responsiveness to Bmp4. Serum-free, defined cultures revealed that, in response to Bmp4, competent epiblast cells uniformly expressed key transcriptional regulators Blimp1 and Prdm14 and acquired germ-cell properties, including genome-wide epigenetic reprogramming, in an orderly fashion. Notably, the induced cells contributed to both spermatogenesis and fertility of offspring. By identifying a signaling principle in germ cell specification, our study establishes a robust strategy for reconstituting the mammalian germ cell lineage in vitro.


Asunto(s)
Proteína Morfogenética Ósea 4/fisiología , Linaje de la Célula/fisiología , Células Germinativas , Transducción de Señal/fisiología , Animales , Proteínas Morfogenéticas Óseas/fisiología , Diferenciación Celular/fisiología , Proteínas de Unión al ADN , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Embrión de Mamíferos/fisiología , Células Germinativas/citología , Células Germinativas/fisiología , Masculino , Mesodermo/citología , Mesodermo/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Proteínas de Unión al ARN , Células Madre/citología , Células Madre/fisiología , Testículo/citología , Testículo/fisiología , Factores de Transcripción/fisiología , Proteínas Wnt/fisiología
8.
Bioorg Med Chem Lett ; 59: 128572, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35066140

RESUMEN

Intestinal sodium-dependent phosphate transport protein 2b (SLC34A2, NaPi2b) inhibitors are expected to be potential new candidates for anti-hyperphosphatemia drugs. However, a risk of on-target side effects based on the inhibition of NaPi2b in the lung and testis has been reported. To identify gut-selective (minimally systemic) NaPi2b inhibitors, we prepared and evaluated 1H-pyrazole-4-carbonyl-4,5,6,7-tetrahydrobenzo[b]thiophene derivatives with highly polar functional groups to reduce systemic exposure. As a result, compounds 36a and 36b showed a good activity in vitro and a low bioavailability in Sprague-Dawley (SD) rats. However, these compounds did not suppress phosphate absorption in SD rats. This lack of in vivo efficacy could be due to the high hydrophobicity of these compounds. The results of further investigations of other classes of compounds with appropriate physical properties will be reported in due course.


Asunto(s)
Diseño de Fármacos , Pirazoles/farmacología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIb/antagonistas & inhibidores , Tiofenos/farmacología , Administración Oral , Animales , Relación Dosis-Respuesta a Droga , Humanos , Inyecciones Intravenosas , Masculino , Estructura Molecular , Pirazoles/administración & dosificación , Pirazoles/química , Ratas , Ratas Sprague-Dawley , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIb/metabolismo , Solubilidad , Relación Estructura-Actividad , Tiofenos/administración & dosificación , Tiofenos/química
9.
Bioorg Med Chem Lett ; 65: 128700, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35346844

RESUMEN

We previously reported thiophene derivatives as gut-selective (minimally systemic) and potent sodium-dependent phosphate transport protein 2b (SLC34A2, NaPi2b) inhibitors. However, these derivatives did not suppress phosphate absorption form the intestinal tract in Sprague-Dawley (SD) rats. The lack of efficacy in vivo could be due to the high hydrophobicity of these compounds. In this report, we identified novel pyridine derivatives as gut-selective NaPi2b inhibitors with good activity in vitro and relatively low hydrophobicity. Especially, gut-selective compound 20b suppressed phosphate absorption in SD rats. These results suggest that physical properties, such as the hydrophobicity of the compounds, might affect the in vivo efficacy.


Asunto(s)
Fosfatos , Piridinas , Animales , Fosfatos/metabolismo , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Tiofenos
10.
Bioorg Med Chem ; 66: 116783, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35576656

RESUMEN

Intestinal sodium-dependent phosphate transport protein 2b (SLC34A2, NaPi2b) inhibitors are expected to be potential new candidates for anti-hyperphosphatemia drugs. However, a risk of on-target side effects based on the inhibition of NaPi2b in the lung and testis has been reported.In this article, we report on our identification of novel indole derivatives as gut-selective NaPi2b inhibitors with good activity, low systemic exposure and moderate hydrophobicity.In particular, gut-selective compound 27, with even lower bioavailability and lower systemic exposure as compared to previously reported pyridine derivatives, demonstrated excellent phosphate absorption-inhibitory effect in SD rats. Compound 27 has an ideal profile and appears to offer promise as a candidate drug for the treatment of hyperphosphatemia, with minimal risk of side effects due to systemic exposure.


Asunto(s)
Inmunoterapia , Fosfatos , Animales , Indoles/farmacología , Masculino , Ratas , Ratas Sprague-Dawley
11.
BMC Pregnancy Childbirth ; 22(1): 825, 2022 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-36348304

RESUMEN

BACKGROUND: Pregnancy discrimination in the workplace is prevalent worldwide. However, few studies have examined the effects of pregnancy discrimination on mothers' perinatal mental health. We aimed to investigate the association between pregnancy discrimination and postpartum depressive symptoms, and the mediation effects of prenatal depressive symptoms on this association. METHODS: Our sample consisted of 285 Japanese women employed during pregnancy who completed a baseline online survey in May 2020 and a follow-up mail survey two months postpartum. Pregnancy discrimination was defined as exposure to any of 16 forms of disadvantageous treatment or harassment related to pregnancy, prohibited by national guidelines. Prenatal (assessed at baseline) and postpartum (assessed at follow-up) depressive symptoms were measured using the Edinburgh Postnatal Depression Scale. Multiple linear regression and mediation analyses were performed overall and stratified by regular (permanent) and non-regular (precarious) employees. RESULTS: Overall, 23.9% of participants experienced pregnancy discrimination during pregnancy. After adjusting for potential confounders, pregnancy discrimination was significantly associated with postpartum depressive symptoms (coefficient 1.76, 95% confidence interval [CI] 0.65-2.88). When stratified by employment type, these effects were observable among non-regular employees (coefficient 2.51, 95% CI 0.45-4.57) but not regular employees. Mediation analysis showed that prenatal depressive symptoms mediated 57.1% (95% CI 20.1-94.1%) of the association between pregnancy discrimination and postpartum depressive symptoms among all participants, with a greater effect among non-regular employees (64.1% [95% CI 18.5-109.8%]). CONCLUSIONS: Pregnancy discrimination has adverse effects on postpartum depressive symptoms, partially through prenatal depressive symptoms, especially among non-regular employees. To prevent perinatal depression in female workers, employers should comply with legislation and take preventive measures against pregnancy discrimination, while considering vulnerable employees.


Asunto(s)
Depresión Posparto , Embarazo , Femenino , Humanos , Depresión Posparto/diagnóstico , Depresión Posparto/epidemiología , Depresión Posparto/psicología , Estudios de Seguimiento , Depresión/diagnóstico , Depresión/epidemiología , Depresión/psicología , Periodo Posparto/psicología , Madres/psicología
12.
EMBO J ; 36(21): 3100-3119, 2017 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-28928204

RESUMEN

The mechanism for sex determination in mammalian germ cells remains unclear. Here, we reconstitute the female sex determination in mouse germ cells in vitro under a defined condition without the use of gonadal somatic cells. We show that retinoic acid (RA) and its key effector, STRA8, are not sufficient to induce the female germ-cell fate. In contrast, bone morphogenetic protein (BMP) and RA synergistically induce primordial germ cells (PGCs)/PGC-like cells (PGCLCs) derived from embryonic stem cells (ESCs) into fetal primary oocytes. The induction is characterized by entry into the meiotic prophase, occurs synchronously and recapitulates cytological and transcriptome progression in vivo faithfully. Importantly, the female germ-cell induction necessitates a proper cellular competence-most typically, DNA demethylation of relevant genes-which is observed in appropriately propagated PGCs/PGCLCs, but not in PGCs/PGCLCs immediately after induction. This provides an explanation for the differential function of BMP signaling between PGC specification and female germ-cell induction. Our findings represent a framework for a comprehensive delineation of the sex-determination pathway in mammalian germ cells, including humans.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Morfogenéticas Óseas/farmacología , Regulación del Desarrollo de la Expresión Génica , Células Madre Embrionarias de Ratones/efectos de los fármacos , Oocitos/efectos de los fármacos , Tretinoina/farmacología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Femenino , Feto , Perfilación de la Expresión Génica , Genes Reporteros , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Endogámicos ICR , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Oocitos/citología , Oocitos/crecimiento & desarrollo , Oocitos/metabolismo , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Profase , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Procesos de Determinación del Sexo , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
13.
EMBO J ; 36(13): 1888-1907, 2017 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-28559416

RESUMEN

The expansion of primordial germ cells (PGCs), the precursors for the oocytes and spermatozoa, is a key challenge in reproductive biology/medicine. Using a chemical screening exploiting PGC-like cells (PGCLCs) induced from mouse embryonic stem cells (ESCs), we here identify key signaling pathways critical for PGCLC proliferation. We show that the combinatorial application of Forskolin and Rolipram, which stimulate cAMP signaling via different mechanisms, expands PGCLCs up to ~50-fold in culture. The expanded PGCLCs maintain robust capacity for spermatogenesis, rescuing the fertility of infertile mice. Strikingly, during expansion, PGCLCs comprehensively erase their DNA methylome, including parental imprints, in a manner that precisely recapitulates genome-wide DNA demethylation in gonadal germ cells, while essentially maintaining their identity as sexually uncommitted PGCs, apparently through appropriate histone modifications. By establishing a paradigm for PGCLC expansion, our system reconstitutes the epigenetic "blank slate" of the germ line, an immediate precursory state for sexually dimorphic differentiation.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Células Madre Embrionarias/fisiología , Epigénesis Genética , Células Germinativas/crecimiento & desarrollo , Animales , Colforsina/metabolismo , Células Germinativas/efectos de los fármacos , Ratones , Rolipram/metabolismo , Transducción de Señal
14.
Biol Reprod ; 104(2): 344-360, 2021 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-33079185

RESUMEN

Primordial germ cells (PGCs) are the founding population of the germ cell lineage that undergo a multistep process to generate spermatozoa or oocytes. Establishing an appropriate culture system for PGCs is a key challenge in reproductive biology. By a chemical screening using mouse PGC-like cells (mPGCLCs), which were induced from mouse embryonic stem cells, we reported previously that forskolin and rolipram synergistically enhanced the proliferation/survival of mPGCLCs with an average expansion rate of ~20-fold. In the present study, we evaluated other chemicals or cytokines to see whether they would improve the current mPGCLC culture system. Among the chemicals and cytokines examined, in the presence of forskolin and rolipram, cyclosporin A (CsA) and fibroblast growth factors (FGFs: FGF2 and FGF10) effectively enhanced the expansion of mPGCLCs in vitro (~50-fold on average). During the expansion by CsA or FGFs, mPGCLCs comprehensively erased their DNA methylation to acquire a profile equivalent to that of gonadal germ cells in vivo, while maintaining their highly motile phenotype as well as their transcriptional properties as sexually uncommitted PGCs. Importantly, these mPGCLCs robustly contributed to spermatogenesis and produced fertile offspring. Furthermore, mouse PGCs (mPGCs) cultured with CsA ex vivo showed transcriptomes and DNA methylomes similar to those of cultured mPGCLCs. The improved culture system for mPGCLCs/mPGCs would be instructive for addressing key questions in PGC biology, including the mechanisms for germ cell migration, epigenetic reprogramming, and sex determination of the germline.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ciclosporina/farmacología , Factor 10 de Crecimiento de Fibroblastos/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Células Germinativas/efectos de los fármacos , Animales , Apoptosis , Ciclo Celular , Proliferación Celular/fisiología , Colforsina/farmacología , Inhibidores Enzimáticos/farmacología , Células Germinativas/fisiología , Ratones , Rolitetraciclina/farmacología , Transducción de Señal/efectos de los fármacos , Secuenciación Completa del Genoma
15.
PLoS Biol ; 16(9): e2004874, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30256801

RESUMEN

Formin is one of the two major classes of actin binding proteins (ABPs) with nucleation and polymerization activity. However, despite advances in our understanding of its biochemical activity, whether and how formins generate specific architecture of the actin cytoskeleton and function in a physiological context in vivo remain largely obscure. It is also unknown how actin filaments generated by formins interact with other ABPs in the cell. Here, we combine genetic manipulation of formins mammalian diaphanous homolog1 (mDia1) and 3 (mDia3) with superresolution microscopy and single-molecule imaging, and show that the formins mDia1 and mDia3 are dominantly expressed in Sertoli cells of mouse seminiferous tubule and together generate a highly dynamic cortical filamentous actin (F-actin) meshwork that is continuous with the contractile actomyosin bundles. Loss of mDia1/3 impaired these F-actin architectures, induced ectopic noncontractile espin1-containing F-actin bundles, and disrupted Sertoli cell-germ cell interaction, resulting in impaired spermatogenesis. These results together demonstrate the previously unsuspected mDia-dependent regulatory mechanism of cortical F-actin that is indispensable for mammalian sperm development and male fertility.


Asunto(s)
Actinas/metabolismo , Proteínas Portadoras/metabolismo , Fertilidad , Células de Sertoli/metabolismo , Espermatogénesis , Actomiosina/metabolismo , Uniones Adherentes/metabolismo , Animales , Adhesión Celular , Células Cultivadas , Forminas , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Polimerizacion , Túbulos Seminíferos/metabolismo , Espermatozoides/citología , Espermatozoides/metabolismo
16.
Mol Cell ; 52(3): 380-92, 2013 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-24120664

RESUMEN

Sox2 is a transcription factor required for the maintenance of pluripotency. It also plays an essential role in different types of multipotent stem cells, raising the possibility that Sox2 governs the common stemness phenotype. Here we show that Sox2 is a critical downstream target of fibroblast growth factor (FGF) signaling, which mediates self-renewal of trophoblast stem cells (TSCs). Sustained expression of Sox2 together with Esrrb or Tfap2c can replace FGF dependency. By comparing genome-wide binding sites of Sox2 in embryonic stem cells (ESCs) and TSCs combined with inducible knockout systems, we found that, despite the common role in safeguarding the stem cell state, Sox2 regulates distinct sets of genes with unique functions in these two different yet developmentally related types of stem cells. Our findings provide insights into the functional versatility of transcription factors during embryogenesis, during which they can be recursively utilized in a variable manner within discrete network structures.


Asunto(s)
Diferenciación Celular/genética , Células Madre Embrionarias/metabolismo , Factores de Transcripción SOXB1/metabolismo , Trofoblastos/metabolismo , Animales , Línea Celular , Desarrollo Embrionario/genética , Células Madre Embrionarias/citología , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Factores de Transcripción SOXB1/inmunología , Transducción de Señal/genética , Células Madre/citología , Células Madre/metabolismo , Factor de Transcripción AP-2/metabolismo , Trofoblastos/citología
17.
Vet Radiol Ultrasound ; 62(4): 483-489, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33855776

RESUMEN

Two-dimensional shear wave elastography (2D-SWE) can be used to quantitatively evaluate the elastic modulus of the liver as shear wave velocity (SWV), which can noninvasively predict clinically relevant hepatic fibrosis in both dogs and humans. However, extrahepatic biliary obstruction (EHBO), regardless of the presence of clinically relevant hepatic fibrosis, can influence SWVs in humans and thus may interfere with hepatic fibrosis prediction using 2D-SWE in dogs. The aim of this prospective, observational, and one-group pretest-posttest study is to investigate whether SWV measured by 2D-SWE displays a difference between dogs with and without EHBO. A total of 20 dogs were included (7 with EHBO and 13 with gallbladder pathology but no EHBO) that underwent preoperative SWV measurement using 2D-SWE. In all dogs, stages of hepatic fibrosis were evaluated histopathologically using a scoring scheme. In addition, postoperative SWVs in dogs with EHBO relieved via laparotomy were also evaluated. The median (range) SWVs in the dogs with and without EHBO were 1.91 (1.81-2.54) m/s and 1.57 (1.37-1.64) m/s, respectively. Although there was no significant difference in the histopathological hepatic fibrosis stages between the dogs with and without EHBO, the preoperative SWVs in the dogs with EHBO were significantly higher than in dogs without EHBO (P = .0004), and SWVs were found to decrease significantly after surgery (P = .0097). This study demonstrates that EHBO can increase the SWV of dogs without clinically relevant hepatic fibrosis and can interfere with the prediction of noninvasive hepatic fibrosis using 2D-SWE.


Asunto(s)
Colestasis , Enfermedades de los Perros , Diagnóstico por Imagen de Elasticidad , Cirrosis Hepática , Animales , Perros , Masculino , Colestasis/complicaciones , Enfermedades de los Perros/diagnóstico por imagen , Enfermedades de los Perros/patología , Diagnóstico por Imagen de Elasticidad/veterinaria , Cirrosis Hepática/complicaciones , Cirrosis Hepática/diagnóstico por imagen , Cirrosis Hepática/patología , Cirrosis Hepática/veterinaria , Estudios Prospectivos
18.
Biol Reprod ; 102(3): 620-638, 2020 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-31724030

RESUMEN

In vitro reconstitution of germ-cell development from pluripotent stem cells (PSCs) has created key opportunities to explore the fundamental mechanisms underlying germ-cell development, particularly in mice and humans. Importantly, such investigations have clarified critical species differences in the mechanisms regulating mouse and human germ-cell development, highlighting the necessity of establishing an in vitro germ-cell development system in other mammals, such as non-human primates. Here, we show that multiple lines of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) in cynomolgus monkeys (Macaca fascicularis; cy) can be maintained stably in an undifferentiated state under a defined condition with an inhibitor for WNT signaling, and such PSCs are induced efficiently into primordial germ cell-like cells (PGCLCs) bearing a transcriptome similar to early cyPGCs. Interestingly, the induction kinetics of cyPGCLCs from cyPSCs is faster than that of human (h) PGCLCs from hPSCs, and while the transcriptome dynamics during cyPGCLC induction is relatively similar to that during hPGCLC induction, it is substantially divergent from that during mouse (m) PGCLC induction. Our findings delineate common as well as species-specific traits for PGC specification, creating a foundation for parallel investigations into the mechanism for germ-cell development in mice, monkeys, and humans.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Pluripotentes/citología , Animales , Células Madre Pluripotentes Inducidas/citología , Macaca fascicularis , Transcriptoma
19.
Nature ; 501(7466): 222-6, 2013 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-23913270

RESUMEN

The germ-cell lineage ensures the continuity of life through the generation of male and female gametes, which unite to form a totipotent zygote. We have previously demonstrated that, by using cytokines, embryonic stem cells and induced pluripotent stem cells can be induced into epiblast-like cells (EpiLCs) and then into primordial germ cell (PGC)-like cells with the capacity for both spermatogenesis and oogenesis, creating an opportunity for understanding and regulating mammalian germ-cell development in both sexes in vitro. Here we show that, without cytokines, simultaneous overexpression of three transcription factors, Blimp1 (also known as Prdm1), Prdm14 and Tfap2c (also known as AP2γ), directs EpiLCs, but not embryonic stem cells, swiftly and efficiently into a PGC state. Notably, Prdm14 alone, but not Blimp1 or Tfap2c, suffices for the induction of the PGC state in EpiLCs. The transcription-factor-induced PGC state, irrespective of the transcription factors used, reconstitutes key transcriptome and epigenetic reprogramming in PGCs, but bypasses a mesodermal program that accompanies PGC or PGC-like-cell specification by cytokines including bone morphogenetic protein 4. Notably, the transcription-factor-induced PGC-like cells contribute to spermatogenesis and fertile offspring. Our findings provide a new insight into the transcriptional logic for PGC specification, and create a foundation for the transcription-factor-based reconstitution and regulation of mammalian gametogenesis.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Células Germinativas/citología , Células Germinativas/metabolismo , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Proteínas de Unión al ADN , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Epigénesis Genética , Femenino , Fertilidad , Perfilación de la Expresión Génica , Estratos Germinativos/citología , Masculino , Mesodermo/citología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Transgénicos , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Proteínas de Unión al ARN , Espermatogénesis , Factor de Transcripción AP-2/genética , Factor de Transcripción AP-2/metabolismo , Factores de Transcripción/genética
20.
Nucleic Acids Res ; 45(21): 12152-12169, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-28981894

RESUMEN

Single transcription factors (TFs) regulate multiple developmental pathways, but the underlying mechanisms remain unclear. Here, we quantitatively characterized the genome-wide occupancy profiles of BLIMP1, a key transcriptional regulator for diverse developmental processes, during the development of three germ-layer derivatives (photoreceptor precursors, embryonic intestinal epithelium and plasmablasts) and the germ cell lineage (primordial germ cells). We identified BLIMP1-binding sites shared among multiple developmental processes, and such sites were highly occupied by BLIMP1 with a stringent recognition motif and were located predominantly in promoter proximities. A subset of bindings common to all the lineages exhibited a new, strong recognition sequence, a GGGAAA repeat. Paradoxically, however, the shared/common bindings had only a slight impact on the associated gene expression. In contrast, BLIMP1 occupied more distal sites in a cell type-specific manner; despite lower occupancy and flexible sequence recognitions, such bindings contributed effectively to the repression of the associated genes. Recognition motifs of other key TFs in BLIMP1-binding sites had little impact on the expression-level changes. These findings suggest that the shared/common sites might serve as potential reservoirs of BLIMP1 that functions at the specific sites, providing the foundation for a unified understanding of the genome regulation by BLIMP1, and, possibly, TFs in general.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Animales , Sitios de Unión , Diferenciación Celular/genética , ADN/química , ADN/metabolismo , Embrión de Mamíferos/metabolismo , Femenino , Técnicas de Sustitución del Gen , Genoma , Células Germinativas/metabolismo , Masculino , Ratones , Motivos de Nucleótidos , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA