Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
EMBO Rep ; 23(12): e55478, 2022 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-36278510

RESUMEN

Tissue stem cells (SCs) divide infrequently as a protective mechanism against internal and external stresses associated with aging. Here, we demonstrate that slow- and fast-cycling SCs in the mouse skin epidermis undergo distinct aging processes. Two years of lineage tracing reveals that Dlx1+ slow-cycling clones expand into the fast-cycling SC territory, while the number of Slc1a3+ fast-cycling clones gradually declines. Transcriptome analysis further indicate that the molecular properties of each SC population are altered with age. Mice lacking fibulin 7, an extracellular matrix (ECM) protein, show early impairments resembling epidermal SC aging, such as the loss of fast-cycling clones, delayed wound healing, and increased expression of inflammation- and differentiation-related genes. Fibulin 7 interacts with structural ECM and matricellular proteins, and the overexpression of fibulin 7 in primary keratinocytes results in slower proliferation and suppresses differentiation. These results suggest that fibulin 7 plays a crucial role in maintaining tissue resilience and epidermal SC heterogeneity during skin aging.


Asunto(s)
Proteínas de Unión al Calcio , Envejecimiento de la Piel , Animales , Ratones , Matriz Extracelular , Envejecimiento de la Piel/genética , Células Madre
2.
J Vis Exp ; (175)2021 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-34633387

RESUMEN

For years, most studies involving keratinocytes have been conducted using human and mouse skin epidermal keratinocytes. Recently, oral keratinocytes have attracted attention because of their unique function and characteristics. They maintain the homeostasis of the oral epithelium and serve as resources for applications in regenerative therapies. However, in vitro studies that use oral primary keratinocytes from adult mice have been limited due to the lack of an efficient and well-established culture protocol. Here, oral primary keratinocytes were isolated from the palate tissues of adult mice and cultured in a commercial low-calcium medium supplemented with a chelexed-serum. Under these conditions, keratinocytes were maintained in a proliferative or stem cell-like state, and their differentiation was inhibited even after increased passages. Marker expression analysis showed that the cultured oral keratinocytes expressed the basal cell markers p63, K14, and α6-integrin and were negative for the differentiation marker K13 and the fibroblast marker PDGFRα. This method produced viable and culturable cells suitable for downstream applications in the study of oral epithelial stem cell functions in vitro.


Asunto(s)
Epidermis , Queratinocitos , Animales , Diferenciación Celular , Ratones , Hueso Paladar , Células Madre
3.
Sci Rep ; 11(1): 8683, 2021 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-33883668

RESUMEN

The maladaptive remodeling of vessel walls with neointima formation is a common feature of proliferative vascular diseases. It has been proposed that neointima formation is caused by the dedifferentiation of mature smooth muscle cells (SMCs). Recent evidence suggests that adventitial cells also participate in neointima formation; however, their cellular dynamics are not fully understood. In this study, we utilized a lineage tracing model of platelet-derived growth factor receptor alpha (PDGFRa) cells and examined cellular behavior during homeostasis and injury response. PDGFRa marked adventitial cells that were largely positive for Sca1 and a portion of medial SMCs, and both cell types were maintained for 2 years. Upon carotid artery ligation, PDGFRa-positive (+) cells were slowly recruited to the neointima and exhibited an immature SMC phenotype. In contrast, in a more severe wire denudation injury, PDGFRa+ cells were recruited to the neointima within 14 days and fully differentiated into SMCs. Under pressure overload induced by transverse aortic constriction, PDGFRa+ cells developed marked adventitial fibrosis. Taken together, our observations suggest that PDGFRa+ cells serve as a reservoir of adventitial cells and a subset of medial SMCs and underscore their context-dependent response to vascular injuries.


Asunto(s)
Vasos Sanguíneos/lesiones , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Adventicia/metabolismo , Animales , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Vasos Sanguíneos/fisiología , Proliferación Celular , Homeostasis , Masculino , Ratones , Ratones Transgénicos , Neointima/metabolismo
4.
Sci Adv ; 7(7)2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33568475

RESUMEN

Stem cell (SC) proliferation and differentiation organize tissue homeostasis. However, how SCs regulate coordinate tissue scaling in dynamic organs remain unknown. Here, we delineate SC regulations in dynamic skin. We found that interfollicular epidermal SCs (IFESCs) shape basal epidermal proliferating clusters (EPCs) in expanding abdominal epidermis of pregnant mice and proliferating plantar epidermis. EPCs consist of IFESC-derived Tbx3+-basal cells (Tbx3+-BCs) and their neighboring cells where Adam8-extracellular signal-regulated kinase signaling is activated. Clonal lineage tracing revealed that Tbx3+-BC clones emerge in the abdominal epidermis during pregnancy, followed by differentiation after parturition. In the plantar epidermis, Tbx3+-BCs are sustained as long-lived SCs to maintain EPCs invariably. We showed that Tbx3+-BCs are vasculature-dependent IFESCs and identified mechanical stretch as an external cue for the vasculature-driven EPC formation. Our results uncover vasculature-mediated IFESC regulations, which explain how the epidermis adjusts its size in orchestration with dermal constituents in dynamic skin.

5.
Development ; 147(24)2020 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-33199446

RESUMEN

Adult tissues contain label-retaining cells (LRCs), which are relatively slow-cycling and considered to represent a property of tissue stem cells (SCs). In the ocular surface epithelium, LRCs are present in the limbus and conjunctival fornix; however, the character of these LRCs remains unclear, owing to lack of appropriate molecular markers. Using three CreER transgenic mouse lines, we demonstrate that the ocular surface epithelium accommodates spatially distinct populations with different cell division dynamics. In the limbus, long-lived Slc1a3CreER-labeled SCs either migrate centripetally toward the central cornea or slowly expand their clones laterally within the limbal region. In the central cornea, non-LRCs labeled with Dlx1CreER and K14CreER behave as short-lived progenitor cells. The conjunctival epithelium in the bulbar, fornix and palpebral compartment is regenerated by regionally unique SC populations. Severe damage to the cornea leads to the cancellation of SC compartments and conjunctivalization, whereas milder limbal injury induces a rapid increase of laterally expanding clones in the limbus. Taken together, our work defines compartmentalized multiple SC/progenitor populations of the mouse eye in homeostasis and their behavioral changes in response to injury.


Asunto(s)
Epitelio Corneal/crecimiento & desarrollo , Transportador 1 de Aminoácidos Excitadores/genética , Proteínas de Homeodominio/genética , Células Madre/citología , Factores de Transcripción/genética , Animales , División Celular/genética , Linaje de la Célula/genética , Células Cultivadas , Conjuntiva/crecimiento & desarrollo , Córnea/crecimiento & desarrollo , Homeostasis/genética , Humanos , Limbo de la Córnea/crecimiento & desarrollo , Ratones , Ratones Transgénicos
6.
Aging Cell ; 19(8): e13190, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32681764

RESUMEN

Aging in the epidermis is marked by a gradual decline in barrier function, impaired wound healing, hair loss, and an increased risk of cancer. This could be due to age-related changes in the properties of epidermal stem cells and defective interactions with their microenvironment. Currently, no biochemical tools are available to detect and evaluate the aging of epidermal stem cells. The cellular glycosylation is involved in cell-cell communications and cell-matrix adhesions in various physiological and pathological conditions. Here, we explored the changes of glycans in epidermal stem cells as a potential biomarker of aging. Using lectin microarray, we performed a comprehensive glycan profiling of freshly isolated epidermal stem cells from young and old mouse skin. Epidermal stem cells exhibited a significant difference in glycan profiles between young and old mice. In particular, the binding of a mannose-binder rHeltuba was decreased in old epidermal stem cells, whereas that of an α2-3Sia-binder rGal8N increased. These glycan changes were accompanied by upregulation of sialyltransferase, St3gal2 and St6gal1 and mannosidase Man1a genes in old epidermal stem cells. The modification of cell surface glycans by overexpressing these glycogenes leads to a defect in the regenerative ability of epidermal stem cells in culture. Hence, our study suggests the age-related global alterations in cellular glycosylation patterns and its potential contribution to the stem cell function. These glycan modifications detected by lectins may serve as molecular markers for aging, and further functional studies will lead us to a better understanding of the process of skin aging.


Asunto(s)
Lectinas/metabolismo , Polisacáridos/metabolismo , Envejecimiento , Animales , Femenino , Glicosilación , Humanos , Masculino , Ratones , Análisis por Micromatrices , Células Madre/metabolismo
7.
Stem Cell Reports ; 14(1): 34-48, 2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31866458

RESUMEN

Epigenetic mechanisms controlling adult mammalian stem cell (SC) dynamics might be critical for tissue regeneration but are poorly understood. Mouse skin and hair follicle SCs (HFSCs) display reduced histone H3 K4me3, K9me3, and K27me3 methylation levels (hypomethylation) preceding hair growth. Chemical inhibition of relevant histone demethylases impairs subsequent differentiation and growth of HFs and delays wound healing. In wounding, this impairs epithelial cell differentiation and blood vessel recruitment, but not proliferation and fibroblast recruitment. With Aspm-CreER as a newfound inter-follicular epidermis lineage-labeling tool, and Lgr5-CreER for hair follicles, we demonstrate a reduced contribution of both lineages to wound healing after interfering with hypomethylation. Blocked hypomethylation increases BMP4 expression and selectively upregulates H3 K4me3 on the Bmp4 promoter, which may explain the effects on HFSC quiescence, hair cycle, and injury repair. Thus, transient hypomethylation of histone H3 K4/9/27me3 is essential for adult skin epithelial SC dynamics for proper tissue homeostasis and repair.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Diferenciación Celular , Histonas/metabolismo , Piel/citología , Piel/metabolismo , Cicatrización de Heridas , Animales , Biomarcadores , Proteínas Morfogenéticas Óseas/metabolismo , Proliferación Celular , Células Epidérmicas/citología , Células Epidérmicas/metabolismo , Inmunohistoquímica , Metilación , Ratones , Transducción de Señal
8.
PLoS One ; 14(5): e0215908, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31091266

RESUMEN

Delayed wound healing and reduced barrier function with an increased risk of cancer are characteristics of aged skin and one possible mechanism is misregulation or dysfunction of epidermal stem cells during aging. Recent studies have identified heterogeneous stem cell populations within the mouse interfollicular epidermis that are defined by territorial distribution and cell division frequency; however, it is unknown whether the individual stem cell populations undergo distinct aging processes. Here we provide comprehensive characterization of age-related changes in the mouse epidermis within the specific territories of slow-cycling and fast-dividing stem cells using old wild-type, senescence-accelerated mouse prone 1 (SAMP1) and SAMP8 mice. During aging, the epidermis exhibits structural changes such as irregular micro-undulations and overall thinning of the tissue. We also find that, in the old epidermis, proliferation is preferentially decreased in the region where fast-dividing stem cells reside whereas the lineage differentiation marker appears to be more affected in the slow-cycling stem cell region. Furthermore, SAMP8, but not SAMP1, exhibits precocious aging similar to that of aged wild-type mice, suggesting a potential use of this model for aging study of the epidermis and its stem cells. Taken together, our study reveals distinct aging processes governing the two epidermal stem cell populations and suggests a potential mechanism in differential responses of compartmentalized stem cells and their niches to aging.


Asunto(s)
Epidermis/metabolismo , Células Madre/citología , Animales , Diferenciación Celular , División Celular , Proliferación Celular , Senescencia Celular , Ratones , Especificidad de la Especie
9.
Matrix Biol ; 74: 5-20, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29730503

RESUMEN

Ectopic calcification occurs during development of chronic kidney disease and has a negative impact on long-term prognosis. The precise molecular mechanism and prevention strategies, however, are not established. Fibulin-7 (Fbln7) is a matricellular protein structurally similar to elastogenic short fibulins, shown to bind dental mesenchymal cells and heparin. Here, we report that Fbln7 is highly expressed in renal tubular epithelium in the adult kidney and mediates renal calcification in mice. In vitro analysis revealed that Fbln7 bound heparin at the N-terminal coiled-coil domain. In Fbln7-expressing CHO-K1 cells, exogenous heparin increased the release of Fbln7 into conditioned media in a dose-dependent manner. This heparin-induced Fbln7 release was abrogated in CHO-745 cells lacking heparan sulfate proteoglycan or in CHO-K1 cells expressing the Fbln7 mutant lacking the N-terminal coiled-coil domain, suggesting that Fbln7 was tethered to pericellular matrix via this domain. Interestingly, Fbln7 knockout (Fbln7-/-) mice were protected from renal tubular calcification induced by high phosphate diet. Mechanistically, Fbln7 bound artificial calcium phosphate particles (aCPP) implicated in calcification and renal inflammation. Binding was decreased significantly in Fbln7-/- primary kidney cells relative to wild-type cells. Further, overexpression of Fbln7 increased binding to aCPP. Addition of heparin reduced binding between aCPP and wild-type cells to levels of Fbln7-/- cells. Taken together, our study suggests that Fbln7 is a local mediator of calcium deposition and that releasing Fbln7 from the cell surface by heparin/heparin derivatives or Fbln7 inhibitory antibodies may provide a novel strategy to prevent ectopic calcification in vivo.


Asunto(s)
Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Heparina/metabolismo , Nefrocalcinosis/metabolismo , Animales , Sitios de Unión , Células CHO , Fosfatos de Calcio/metabolismo , Proteínas de Unión al Calcio/química , Membrana Celular/metabolismo , Cricetulus , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Células HEK293 , Proteoglicanos de Heparán Sulfato/metabolismo , Humanos , Ratones , Mutación , Nefrocalcinosis/inducido químicamente , Nefrocalcinosis/genética , Unión Proteica
11.
Nat Cell Biol ; 18(6): 619-31, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27183471

RESUMEN

The interfollicular epidermis regenerates from heterogeneous basal skin cell populations that divide at different rates. It has previously been presumed that infrequently dividing basal cells known as label-retaining cells (LRCs) are stem cells, whereas non-LRCs are short-lived progenitors. Here we employ the H2B-GFP pulse-chase system in adult mouse skin and find that epidermal LRCs and non-LRCs are molecularly distinct and can be differentiated by Dlx1(CreER) and Slc1a3(CreER) genetic marking, respectively. Long-term lineage tracing and mathematical modelling of H2B-GFP dilution data show that LRCs and non-LRCs constitute two distinct stem cell populations with different patterns of proliferation, differentiation and upward cellular transport. During homeostasis, these populations are enriched in spatially distinct skin territories and can preferentially produce unique differentiated lineages. On wounding or selective killing, they can temporarily replenish each other's territory. These two discrete interfollicular stem cell populations are functionally interchangeable and intrinsically well adapted to thrive in distinct skin environments.


Asunto(s)
Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Proliferación Celular/fisiología , Células Epidérmicas , Piel/citología , Células Madre/citología , Envejecimiento , Animales , Diferenciación Celular/genética , Proliferación Celular/genética , Perfilación de la Expresión Génica/métodos , Homeostasis/fisiología , Ratones , Coloración y Etiquetado/métodos
12.
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
13.
Cell Rep ; 6(3): 499-513, 2014 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-24462289

RESUMEN

Quiescent hair follicle (HF) bulge stem cells (SCs) differentiate to early progenitor (EP) hair germ (HG) cells, which divide to produce transit-amplifying matrix cells. EPs can revert to SCs upon injury, but whether this dedifferentiation occurs in normal HF homeostasis (hair cycle) and the mechanisms regulating both differentiation and dedifferentiation are unclear. Here, we use lineage tracing, gain of function, transcriptional profiling, and functional assays to examine the role of observed endogenous Runx1 level changes in the hair cycle. We find that forced Runx1 expression induces hair degeneration (catagen) and simultaneously promotes changes in the quiescent bulge SC transcriptome toward a cell state resembling the EP HG fate. This cell-state transition is functionally reversible. We propose that SC differentiation and dedifferentiation are likely to occur during normal HF degeneration and niche restructuring in response to changes in endogenous Runx1 levels associated with SC location with respect to the niche.


Asunto(s)
Ciclo Celular , Diferenciación Celular , Folículo Piloso/citología , Células Madre/citología , Células Madre/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Perfilación de la Expresión Génica , Folículo Piloso/efectos de los fármacos , Folículo Piloso/crecimiento & desarrollo , Homeostasis/efectos de los fármacos , Homeostasis/genética , Ratones , Ratones Transgénicos , Modelos Biológicos , Células Madre/efectos de los fármacos , Tetraciclina/farmacología , Factores de Tiempo
14.
Int Rev Cell Mol Biol ; 300: 1-50, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23273858

RESUMEN

Stem cells can self-renew and differentiate over extended periods of time. Understanding how stem cells acquire their fates is a central question in stem cell biology. Early work in Drosophila germ line and neuroblast showed that fate choice is achieved by strict asymmetric divisions that can generate each time one stem and one differentiated cell. More recent work suggests that during homeostasis, some stem cells can divide symmetrically to generate two differentiated cells or two identical stem cells to compensate for stem cell loss that occurred by direct differentiation or apoptosis. The interplay of all these factors ensures constant tissue regeneration and the maintenance of stem cell pool size. This interplay can be modeled as a population-deterministic dynamics that, at least in some systems, may be described as stochastic behavior. Here, we overview recent progress made on the characterization of stem cell dynamics in regenerative tissues.


Asunto(s)
Regeneración/fisiología , Células Madre/citología , Células Madre/fisiología , Animales , Caenorhabditis elegans , Diferenciación Celular/fisiología , División Celular/fisiología , Drosophila , Femenino , Humanos , Masculino , Modelos Biológicos , Nicho de Células Madre/fisiología
15.
Stem Cells ; 30(2): 280-91, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22102605

RESUMEN

Stem cells are maintained by both stem cell-extrinsic niche signals and stem cell-intrinsic factors. During murine spermatogenesis, glial cell line-derived neurotrophic factor (GDNF) signal emanated from Sertoli cells and germ cell-intrinsic factor NANOS2 represent key regulators for the maintenance of spermatogonial stem cells. However, it remains unclear how these factors intersect in stem cells to control their cellular state. Here, we show that GDNF signaling is essential to maintain NANOS2 expression, and overexpression of Nanos2 can alleviate the stem cell loss phenotype caused by the depletion of Gfra1, a receptor for GDNF. By using an inducible Cre-loxP system, we show that NANOS2 expression is downregulated upon the conditional knockout (cKO) of Gfra1, while ectopic expression of Nanos2 in GFRA1-negative spermatogonia does not induce de novo GFRA1 expression. Furthermore, overexpression of Nanos2 in the Gfra1-cKO testes prevents precocious differentiation of the Gfra1-knockout stem cells and partially rescues the stem cell loss phenotypes of Gfra1-deficient mice, indicating that the stem cell differentiation can be suppressed by NANOS2 even in the absence of GDNF signaling. Taken together, we suggest that NANOS2 acts downstream of GDNF signaling to maintain undifferentiated state of spermatogonial stem cells.


Asunto(s)
Proteínas Portadoras/fisiología , Diferenciación Celular , Factores de Crecimiento Nervioso/metabolismo , Neuroglía/metabolismo , Transducción de Señal , Espermatogonias/fisiología , Animales , Apoptosis , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proliferación Celular , Células Cultivadas , Femenino , Regulación de la Expresión Génica , Genes Reporteros , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Proteínas Luminiscentes/biosíntesis , Proteínas Luminiscentes/genética , Masculino , Ratones , Ratones Transgénicos , Proteínas de Unión al ARN , Espermatogonias/metabolismo , Testículo/citología , Testículo/metabolismo
16.
PLoS One ; 5(2): e9300, 2010 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-20174582

RESUMEN

BACKGROUND: The regulation of gene expression via a 3' untranslated region (UTR) plays essential roles in the discrimination of the germ cell lineage from somatic cells during embryogenesis. This is fundamental to the continuation of a species. Mouse NANOS3 is an essential protein required for the germ cell maintenance and is specifically expressed in these cells. However, the regulatory mechanisms that restrict the expression of this gene in the germ cells is largely unknown at present. METHODOLOGY/PRINCIPAL FINDINGS: In our current study, we show that differences in the stability of Nanos3 mRNA between germ cells and somatic cells is brought about in a 3'UTR-dependent manner in mouse embryos. Although Nanos3 is transcribed in both cell lineages, it is efficiently translated only in the germ lineage. We also find that the translational suppression of NANOS3 in somatic cells is caused by a 3'UTR-mediated mRNA destabilizing mechanism. Surprisingly, even when under the control of the CAG promoter which induces strong ubiquitous transcription in both germ cells and somatic cells, the addition of the Nanos3-3'UTR sequence to the coding region of exogenous gene was effective in restricting protein expression in germ cells. CONCLUSIONS/SIGNIFICANCE: Our current study thus suggests that Nanos3-3'UTR has an essential role in translational control in the mouse embryo.


Asunto(s)
Regiones no Traducidas 3'/genética , Embrión de Mamíferos/metabolismo , Perfilación de la Expresión Génica , Proteínas de Unión al ARN/genética , Animales , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Femenino , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Proteínas de Unión al ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Proteína Fluorescente Roja
17.
Dev Biol ; 336(2): 222-31, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19818747

RESUMEN

Spermatogonial stem cells (SSCs) reside in undifferentiated type-A spermatogonia and contribute to continuous spermatogenesis by maintaining the balance between self-renewal and differentiation, thereby meeting the biological demand in the testis. Spermatogonia have to date been characterized principally through their morphology, but we herein report the detailed characterization of undifferentiated spermatogonia in mouse testes based on their gene expression profiles in combination with topological features. The detection of the germ cell-specific proteins Nanos2 and Nanos3 as markers of spermatogonia has enabled the clear dissection of complex populations of these cells as Nanos2 was recently shown to be involved in the maintenance of stem cells. Nanos2 is found to be almost exclusively expressed in A(s) to A(pr) cells, whereas Nanos3 is detectable in most undifferentiated spermatogonia (A(s) to A(al)) and differentiating A(1) spermatogonia. In our present study, we find that A(s) and A(pr) can be basically classified into three categories: (1) GFRalpha1(+)Nanos2(+)Nanos3(-)Ngn3(-), (2) GFRalpha1(+)Nanos2(+)Nanos3(+)Ngn3(-), and (3) GFRalpha1(-)Nanos2(+/-)Nanos3(+)Ngn3(+). We propose that the first of these groups is most likely to include the stem cell population and that Nanos3 may function in transit amplifying cells.


Asunto(s)
Biomarcadores/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Unión al ARN/metabolismo , Espermatogonias/metabolismo , Espermatozoides/metabolismo , Animales , Secuencia de Bases , Cartilla de ADN , Femenino , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa
18.
Science ; 325(5946): 1394-8, 2009 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-19745153

RESUMEN

Stem cells give rise to differentiated cell types but also preserve their undifferentiated state through cell self-renewal. With the use of transgenic mice, we found that the RNA-binding protein NANOS2 is essential for maintaining spermatogonial stem cells. Lineage-tracing analyses revealed that undifferentiated spermatogonia expressing Nanos2 self-renew and generate the entire spermatogenic cell lineage. Conditional disruption of postnatal Nanos2 depleted spermatogonial stem cell reserves, whereas mouse testes in which Nanos2 had been overexpressed accumulated spermatogonia with undifferentiated, stem cell-like properties. Thus, NANOS2 is a key stem cell regulator that is expressed in self-renewing spermatogonial stem cells and maintains the stem cell state during murine spermatogenesis.


Asunto(s)
Proteínas Portadoras/metabolismo , Espermatogénesis , Espermatogonias/citología , Células Madre/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas Portadoras/genética , Diferenciación Celular , Linaje de la Célula , Técnicas de Inactivación de Genes , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Proteínas de Unión al ARN/metabolismo , Espermatogonias/metabolismo , Células Madre/metabolismo , Testículo/citología , Testículo/metabolismo , Dedos de Zinc
19.
Development ; 133(21): 4233-43, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17021047

RESUMEN

The expression of C/EBPalpha, which may govern transcription of mature hepatocyte marker genes, was suppressed in periportal hepatoblasts in mouse liver development, leading to biliary cell differentiation. This study was undertaken to analyze how inactivation of the Cebpa gene affects biliary cell differentiation and gene expression of the regulatory genes for that differentiation, including Hnf1b and Hnf6. In the knockout mouse liver at midgestation stages, pseudoglandular structures were abundantly induced in the parenchyma with elevated expression of Hnf6 and Hnf1b mRNAs. The wild-type liver parenchyma expressed mRNAs of these transcription factors at low levels, though periportal biliary progenitors had strong expression of them. These results suggest that expression of Hnf6 and Hnf1b is downstream of C/EBPalpha action in fetal liver development, and that the suppression of C/EBPalpha expression in periportal hepatoblasts may lead to expression of Hnf6 and Hnf1b mRNAs. Immunohistochemical studies with biliary cell markers in knockout livers demonstrated that differentiated biliary epithelial cells were confined to around the portal veins. The suppression of C/EBPalpha expression may result in upregulation of Hnf6 and Hnf1b gene expression, but be insufficient for biliary cell differentiation. When liver fragments of Cebpa-knockout fetuses, in which hepatoblasts were contained as an endodermal component, were transplanted in the testis of Scid (Prkdc) male mice, almost all hepatoblasts gave rise to biliary epithelial cells. Wild-type hepatoblasts constructed mature hepatic tissue accompanied by biliary cell differentiation. These results also demonstrate that the suppression of C/EBPalpha expression may stimulate biliary cell differentiation.


Asunto(s)
Conductos Biliares/citología , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Factor Nuclear 1-beta del Hepatocito/metabolismo , Factor Nuclear 6 del Hepatocito/metabolismo , Hepatocitos/fisiología , Animales , Conductos Biliares/crecimiento & desarrollo , Biomarcadores/metabolismo , Proteína alfa Potenciadora de Unión a CCAAT/genética , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Moléculas de Adhesión Celular/metabolismo , Trasplante de Células , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 1-beta del Hepatocito/genética , Factor Nuclear 6 del Hepatocito/genética , Hepatocitos/citología , Hibridación in Situ , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Hígado/citología , Hígado/crecimiento & desarrollo , Hígado/metabolismo , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Ratones SCID , ARN Mensajero/metabolismo , Receptor Notch2/genética , Receptor Notch2/metabolismo , Proteínas Serrate-Jagged , Testículo/citología , Testículo/embriología , Factor de Crecimiento Transformador beta1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...