Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Dev Biol ; 357(1): 202-10, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21723859

RESUMEN

Understanding how stem cells are maintained in their microenvironment (the niche) is vital for their application in regenerative medicine. Studies of Drosophila male germline stem cells (GSCs) have served as a paradigm in niche-stem cell biology. It is known that the BMP and JAK-STAT pathways are necessary for the maintenance of GSCs in the testis (Kawase et al., 2004; Kiger et al., 2001; Schulz et al., 2004; Shivdasani and Ingham, 2003; Tulina and Matunis, 2001). However, our recent work strongly suggests that BMP signaling is the primary pathway leading to GSC self-renewal (Leatherman and DiNardo, 2010). Here we show that magu controls GSC maintenance by modulating the BMP pathway. We found that magu was specifically expressed from hub cells, and accumulated at the testis tip. Testes from magu mutants exhibited a reduced number of GSCs, yet maintained a normal population of somatic stem cells and hub cells. Additionally, BMP pathway activity was reduced, whereas JAK-STAT activation was retained in mutant testes. Finally, GSC loss caused by the magu mutation could be suppressed by overactivating the BMP pathway in the germline.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Proteínas de Drosophila/fisiología , Drosophila/metabolismo , Proteínas de la Matriz Extracelular/fisiología , Transducción de Señal , Nicho de Células Madre/citología , Testículo/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas Morfogenéticas Óseas/genética , Drosophila/citología , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Embrión no Mamífero/metabolismo , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Células Germinativas/citología , Células Germinativas/metabolismo , Masculino , Mutación , Nicho de Células Madre/metabolismo , Testículo/citología
3.
Nature ; 475(7356): 381-5, 2011 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-21776083

RESUMEN

The gene for the atypical NOTCH ligand delta-like homologue 1 (Dlk1) encodes membrane-bound and secreted isoforms that function in several developmental processes in vitro and in vivo. Dlk1, a member of a cluster of imprinted genes, is expressed from the paternally inherited chromosome. Here we show that mice that are deficient in Dlk1 have defects in postnatal neurogenesis in the subventricular zone: a developmental continuum that results in depletion of mature neurons in the olfactory bulb. We show that DLK1 is secreted by niche astrocytes, whereas its membrane-bound isoform is present in neural stem cells (NSCs) and is required for the inductive effect of secreted DLK1 on self-renewal. Notably, we find that there is a requirement for Dlk1 to be expressed from both maternally and paternally inherited chromosomes. Selective absence of Dlk1 imprinting in both NSCs and niche astrocytes is associated with postnatal acquisition of DNA methylation at the germ-line-derived imprinting control region. The results emphasize molecular relationships between NSCs and the niche astrocyte cells of the microenvironment, identifying a signalling system encoded by a single gene that functions coordinately in both cell types. The modulation of genomic imprinting in a stem-cell environment adds a new level of epigenetic regulation to the establishment and maintenance of the niche, raising wider questions about the adaptability, function and evolution of imprinting in specific developmental contexts.


Asunto(s)
Animales Recién Nacidos/metabolismo , Astrocitos/metabolismo , Impresión Genómica , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis , Nicho de Células Madre/citología , Envejecimiento/genética , Animales , Secuencia de Bases , Proteínas de Unión al Calcio , Membrana Celular/metabolismo , Células Cultivadas , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Femenino , Genotipo , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Bulbo Olfatorio/citología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Nicho de Células Madre/metabolismo
4.
Dev Cell ; 21(1): 159-71, 2011 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-21763616

RESUMEN

The past decade of research on Drosophila stem cells and niches has provided key insights. Fly stem cells do not occupy a special "state" based on novel "stem cell genes" but resemble transiently arrested tissue progenitors. Moreover, individual stem cells and downstream progenitors are highly dynamic and dispensable, not tissue bulwarks. Niches, rather than fixed cell lineages, ensure tissue health by holding stem cells and repressing cell differentiation inside, but not outside. We review the five best-understood adult Drosophila stem cells and argue that the fundamental biology of stem cells and niches is conserved between Drosophila and mice.


Asunto(s)
Drosophila/citología , Nicho de Células Madre/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Drosophila/metabolismo , Nicho de Células Madre/citología
5.
Exp Hematol ; 39(7): 723-9, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21624427

RESUMEN

Several stem cell mobilization strategies have been employed in the past 2 decades, including chemotherapy, hematopoietic growth factors, and chemotherapy plus growth factors. Granulocyte colony-stimulating factor (G-CSF) and granulocyte-macrophage CSF are standard agents approved for peripheral blood stem cell mobilization since the early 1990s. Between 5% and 20% of patients, however, fail to mobilize a sufficient numbers of peripheral blood stem cells in response to G-CSF with or without chemotherapy. Recent advances in defining the basic mechanisms regulating the interactions between hematopoietic stem cells and their marrow niche had led to the discovery that CXCR4 and stromal-cell-derived factor 1α axis play a significant role. Plerixafor, an antagonist of the CXCR4-stromal-cell-derived factor 1α axis has been shown to result in a significant mobilization of hematopoietic stem cells. Numerous clinical trials have demonstrated that the combination of G-CSF and AMD3100 (G+A) resulted in a significant increase in CD34(+) cell yield as compared to the administration of G-CSF alone. In particular, the progenitors mobilized have been shown to comprise a significantly higher proportion of primitive and possibly more potent CD34(+)/CD38(-) subpopulation. Transplantation of PBSC mobilized by G+A administration have led to a rapid and sustained neutrophil and platelet engraftment. Another prospective role of this new class of agents might lie in the mobilization of dormant leukemia stem cells that are well protected by the niche. The future role of CXCR4 antagonists in treatment of hematologic malignancies includes mobilization of hematopoietic stem cells for transplantation and mobilization of leukemia-initiating cells for long-term cure.


Asunto(s)
Células de la Médula Ósea/citología , Movilización de Célula Madre Hematopoyética/métodos , Trasplante de Células Madre de Sangre Periférica/métodos , Nicho de Células Madre/citología , Bencilaminas , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Quimiocina CXCL12/metabolismo , Ciclamas , Sinergismo Farmacológico , Factor Estimulante de Colonias de Granulocitos/farmacología , Compuestos Heterocíclicos/farmacología , Humanos , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/metabolismo , Nicho de Células Madre/efectos de los fármacos , Nicho de Células Madre/metabolismo
6.
PLoS One ; 6(5): e20364, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21647226

RESUMEN

Hematopoietic stem cell (HSC) homeostasis in the adult bone marrow (BM) is regulated by both intrinsic gene expression products and interactions with extrinsic factors in the HSC niche. GRP94, an endoplasmic reticulum chaperone, has been reported to be essential for the expression of specific integrins and to selectively regulate early T and B lymphopoiesis. In GRP94 deficient BM chimeras, multipotent hematopoietic progenitors persisted and even increased, however, the mechanism is not well understood. Here we employed a conditional knockout (KO) strategy to acutely eliminate GRP94 in the hematopoietic system. We observed an increase in HSCs and granulocyte-monocyte progenitors in the Grp94 KO BM, correlating with an increased number of colony forming units. Cell cycle analysis revealed that a loss of quiescence and an increase in proliferation led to an increase in Grp94 KO HSCs. This expansion of the HSC pool can be attributed to the impaired interaction of HSCs with the niche, evidenced by enhanced HSC mobilization and severely compromised homing and lodging ability of primitive hematopoietic cells. Transplanting wild-type (WT) hematopoietic cells into a GRP94 null microenvironment yielded a normal hematology profile and comparable numbers of HSCs as compared to WT control, suggesting that GRP94 in HSCs, but not niche cells, is required for maintaining HSC homeostasis. Investigating this, we further determined that there was a near complete loss of integrin α4 expression on the cell surface of Grp94 KO HSCs, which showed impaired binding with fibronectin, an extracellular matrix molecule known to play a role in mediating HSC-niche interactions. Furthermore, the Grp94 KO mice displayed altered myeloid and lymphoid differentiation. Collectively, our studies establish GRP94 as a novel cell intrinsic factor required to maintain the interaction of HSCs with their niche, and thus regulate their physiology.


Asunto(s)
Células de la Médula Ósea/citología , Comunicación Celular , Retículo Endoplásmico/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Proteínas de la Membrana/metabolismo , Nicho de Células Madre/citología , Animales , Comunicación Celular/genética , Diferenciación Celular/genética , Movimiento Celular/genética , Femenino , Fibronectinas/metabolismo , Regulación de la Expresión Génica/genética , Técnicas de Inactivación de Genes , Proteínas HSP70 de Choque Térmico/deficiencia , Proteínas HSP70 de Choque Térmico/genética , Homeostasis/genética , Integrina alfa4/metabolismo , Linfocitos/citología , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Células Mieloides/citología , Fase de Descanso del Ciclo Celular/genética , Nicho de Células Madre/metabolismo
7.
Blood ; 118(6): 1534-43, 2011 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-21670467

RESUMEN

Hypoxia is emerging as an important characteristic of the hematopoietic stem cell (HSC) niche, but the molecular mechanisms contributing to quiescence, self-renewal, and survival remain elusive. Vascular endothelial growth factor A (VEGFA) is a key regulator of angiogenesis and hematopoiesis. Its expression is commonly regulated by hypoxia-inducible factors (HIF) that are functionally induced in low-oxygen conditions and that activate transcription by binding to hypoxia-response elements (HRE). Vegfa is indispensable for HSC survival, mediated by a cell-intrinsic, autocrine mechanism. We hypothesized that a hypoxic HSC microenvironment is required for maintenance or up-regulation of Vegfa expression in HSCs and therefore crucial for HSC survival. We have tested this hypothesis in the mouse model Vegfa(δ/δ), where the HRE in the Vegfa promoter is mutated, preventing HIF binding. Vegfa expression was reduced in highly purified HSCs from Vegfa(δ/δ) mice, showing that HSCs reside in hypoxic areas. Loss of hypoxia-regulated Vegfa expression increases the numbers of phenotypically defined hematopoietic stem and progenitor cells. However, HSC function was clearly impaired when assessed in competitive transplantation assays. Our data provide further evidence that HSCs reside in a hypoxic microenvironment and demonstrate a novel way in which the hypoxic niche affects HSC fate, via the hypoxia-VEGFA axis.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Oxígeno/metabolismo , Nicho de Células Madre/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Hipoxia de la Célula , Células Cultivadas , Femenino , Citometría de Flujo , Expresión Génica , Genotipo , Placa de Crecimiento/irrigación sanguínea , Placa de Crecimiento/crecimiento & desarrollo , Hematopoyesis/genética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Hexoquinasa/genética , Hexoquinasa/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hígado/citología , Hígado/embriología , Hígado/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfoglicerato Quinasa/genética , Fosfoglicerato Quinasa/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Nicho de Células Madre/citología , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
Stem Cells ; 29(6): 907-12, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21557389

RESUMEN

The adult mammalian central nervous system (CNS) lacks the capacity for regeneration, making it a highly sought-after topic for researchers. The identification of neural stem cells (NSCs) in the adult CNS wiped out a long-held dogma that the adult brain contains a set number of neurons and is incapable of replacing them. The discovery of adult NSCs (aNSCs) stoked the fire for researchers who dream of brain self-repair. Unfortunately, the quiescent nature and limited plasticity of aNSCs diminish their regenerative potential. Recent studies evaluating aNSC plasticity under pathological conditions indicate that a switch from quiescent to active aNSCs in neurogenic regions plays an important role in both repairing the damaged tissue and preserving progenitor pools. Here, we summarize the most recent findings and present questions about characterizing the active and quiescent aNSCs in major neurogenic regions, and factors for maintaining their active and quiescent states, hoping to outline an emerging view for promoting the endogenous aNSC-based regeneration.


Asunto(s)
Células Madre Adultas/fisiología , Proliferación Celular , Senescencia Celular , Células-Madre Neurales/fisiología , Animales , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular , Hipocampo/citología , Hipocampo/fisiología , Humanos , Ventrículos Laterales/citología , Ventrículos Laterales/fisiología , Mucosa Olfatoria/citología , Mucosa Olfatoria/fisiología , Nicho de Células Madre/metabolismo
9.
EMBO Rep ; 12(6): 519-26, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21546910

RESUMEN

Drosophila ovarian germline stem cells (GSCs) are maintained by the extracellular BMP2/4 orthologue Dpp, which is produced from the surrounding somatic niche. The Dpp signal has a short range; it induces a response in GSCs within the niche, but is rapidly extinguished in their progeny only one cell-diameter away. To ensure the correct balance between stem-cell maintenance and differentiation, several regulatory mechanisms that modulate the Dpp signal at many stages of the pathway have been described. Here, we discuss the nature of the ovarian Dpp signal and review the catalogue of mechanisms that regulate it, demonstrating how the exquisite modulation of Dpp signalling in this context can result in precise and robust control of stem-cell fate. This modulation is applicable to other stem-cell environments that use BMPs as a niche signal, and the regulatory mechanisms are conceptually relevant to several other stem-cell systems.


Asunto(s)
Drosophila/metabolismo , Transducción de Señal , Nicho de Células Madre/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Drosophila/citología , Espacio Extracelular/metabolismo , Femenino , Células Germinativas/metabolismo , Humanos , Espacio Intracelular/metabolismo , Ovario/citología , Ovario/metabolismo
10.
J Exp Zool B Mol Dev Evol ; 316(5): 359-70, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21455945

RESUMEN

Antler development is triggered by interactions between antler stem cells resident in the antlerogenic periosteum (AP) and the niche cells in the upper portion of overlying skin mediated by diffusible molecules. These interactive cell populations are interposed by the lower portion of the skin and the subcutaneous loose connective tissue (SLCT). It is known that mechanical deletion of just the central AP (having an area equivalent to the size of a pedicle base) by cutting through the skin and SLCT effectively stimulates the marginal AP to initiate antler development. This study was designed to investigate whether the SLCT layer plays a role in antler development by acting as a physical barrier. The results showed that the marginal AP failed to give rise to an antler after the central AP was cryosurgically destroyed with the preservation of the collagen structure of the SLCT. Furthermore, antler development was significantly advanced when the collagen structures of the skin and SLCT layers were substantially attenuated by repeated sprays with liquid nitrogen while keeping the central AP intact. Therefore, we conclude that the interposing SLCT layer acts as a physical barrier between antler stem cells and the niche cell types, and that timing of antler development is primarily controlled by the permeability of the SLCT layer to the putative interactive diffusible molecules.


Asunto(s)
Cuernos de Venado/crecimiento & desarrollo , Cuernos de Venado/cirugía , Criocirugía/métodos , Periostio/metabolismo , Periostio/cirugía , Piel/metabolismo , Tejido Subcutáneo/metabolismo , Animales , Ciervos , Femenino , Masculino , Osteogénesis , Nicho de Células Madre/metabolismo
11.
Cell Mol Life Sci ; 68(15): 2513-23, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21509540

RESUMEN

The gastrointestinal epithelium is a highly organised tissue that is constantly being renewed. In order to maintain homeostasis, the balance between intestinal stem cell (ISC) self-renewal and differentiation must be carefully regulated. In this review, we describe how the intestinal stem cell niche provides a unique environment to regulate self-renewal and differentiation of ISCs. It has traditionally been believed that the mesenchymal myofibroblasts play an important role in the crosstalk between ISCs and the niche. However, recent evidence in Drosophila and in vertebrates suggests that epithelial cells also contribute to the niche. We discuss the multiple signalling pathways that are utilised to regulate stemness within the niche, including members of the Wnt, BMP and Hedgehog pathways, and how aberrations in these signals lead to disruption of the normal crypt-villus axis. Finally, we also discuss how CDX1 and inhibition of the Notch pathway are important in specifying enterocyte and goblet cell differentiation respectively.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Intestinos/citología , Nicho de Células Madre/fisiología , Animales , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/fisiología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/fisiología , Humanos , Mucosa Intestinal/metabolismo , Intestinos/fisiología , Modelos Biológicos , Nicho de Células Madre/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteínas Wnt/fisiología
12.
Blood ; 117(24): 6552-61, 2011 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-21511956

RESUMEN

In the bone marrow (BM), stromal cells constitute a supportive tissue indispensable for the generation of pro-B/pre-BI, pre-BII, and immature B lymphocytes. IL-7-producing stromal cells constitute a cellular niche for pro-B/pre-BI cells, but no specific stromal cell microenvironment was identified for pre-BII cells expressing a functional pre-B cell receptor (pre-BCR). However expression of the pre-BCR represents a crucial checkpoint during B-cell development. We recently demonstrated that the stromal cell derived-galectin1 (GAL1) is a ligand for the pre-BCR, involved in the proliferation and differentiation of normal mouse pre-BII cells. Here we show that nonhematopoietic osteoblasts and reticular cells in the BM express GAL1. We observed that pre-BII cells, unlike the other B-cell subsets, were specifically localized in close contact with GAL1(+) reticular cells. We also determined that IL-7(+) and GAL1(+) cells represent 2 distinct mesenchymal populations with different BM localization. These results demonstrate the existence of a pre-BII specific stromal cell niche and indicate that early B cells move from IL-7(+) to GAL1(+) supportive BM niches during their development.


Asunto(s)
Médula Ósea , Galectina 1/metabolismo , Células Precursoras de Linfocitos B/fisiología , Nicho de Células Madre/fisiología , Células del Estroma/fisiología , Animales , Médula Ósea/metabolismo , Médula Ósea/fisiología , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/fisiología , Diferenciación Celular/inmunología , Células Cultivadas , Proteínas Fluorescentes Verdes/genética , Interleucina-7/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Células Precursoras de Linfocitos B/metabolismo , Células Precursoras de Linfocitos B/citología , Células Precursoras de Linfocitos B/metabolismo , Nicho de Células Madre/citología , Nicho de Células Madre/metabolismo , Células del Estroma/citología , Células del Estroma/metabolismo
13.
Haematologica ; 96(7): 1041-8, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21459792

RESUMEN

The field of hematopoietic oncology has traditionally focused on the study of hematopoietic cell autonomous genetic events in an effort to understand malignant transformation and develop therapeutics. Although highly rewarding in both aspects, this cell autonomous approach has failed to fully satisfy our need to understand tumor cell behavior and related clinical observations. In recent years, it has been increasingly recognized that the tumor microenvironment plays a pivotal role in cancer initiation and progression. This review will discuss recent experimental evidence in support of this view derived from investigations in both epithelial and hematopoietic systems. Based on this, conceptual views and therapeutic implications will be provided on the emerging role of the bone marrow microenvironment in leukemogenesis.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Sistema Hematopoyético/metabolismo , Nicho de Células Madre/metabolismo , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Regulación Neoplásica de la Expresión Génica , Sistema Hematopoyético/patología , Humanos , Leucemia/metabolismo , Leucemia/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Nicho de Células Madre/patología , Microambiente Tumoral/genética
14.
Plant Signal Behav ; 6(4): 544-6, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21406977

RESUMEN

Shoot apical meristem (SAM) stem cell niche is an interconnected network of distinct cell types; the central zone (CZ) harbors a small pool of stem cells, the stem cell progeny are displaced into the adjacent peripheral zone (PZ) and the rib zone (RZ) located beneath the CZ where they differentiate. Relative ratios of cell types remain constant. Genetic studies have shown that the levels or spatial confinement of WUS, a homeodomain transcription factor to few cells in the RZ is critical for regulating stem cell number. However, static analyses of terminal mutant phenotypes have not revealed WUS-mediated mechanisms of stem cell homeostasis. In a recent study we have employed transient manipulation of WUS levels and live imaging to show that it controls several interdependent processes such as regulation of stem cell number, cell division rates of stem cell progenitors and their patterns of differentiation, thus providing robustness to the process.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Proteínas de Homeodominio/metabolismo , Meristema/citología , Brotes de la Planta/citología , Nicho de Células Madre/citología , Nicho de Células Madre/metabolismo , Células Madre/citología , Células Madre/metabolismo , Arabidopsis/citología , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Regulación de la Expresión Génica de las Plantas , Proteínas de Homeodominio/genética , Modelos Biológicos
15.
Tissue Eng Part A ; 17(11-12): 1651-61, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21306280

RESUMEN

After the clinical insertion of a bone biomaterial, the surrounding osteoblasts would migrate and attach to the implant surface and foster a microenvironment that largely determines the differentiation fate of the comigrated mesenchymal stem cells. Whether the fostered microenvironment is suitable for osteogenic differentiation of mesenchymal stem cells is critical for the subsequent osseointegration. In this study, we determined (1) how the spherical or rod-shaped hydroxyapatite nanoparticles (nHA) incorporated poly(ɛ-caprolactone) (PCL) films (PCL-spherical nHA, PCL-rod nHA) interact with primary human osteoblasts (HOBs); (2) how the microenvironment rendered by their interaction affects osteogenic differentiation of adipose tissue-derived mesenchymal stem cells (ASCs). HOBs were seeded on PCL, PCL-spherical nHA, and PCL-rod nHA films, respectively. When cultured alone, the HOBs on PCL-rod nHA films showed most efficient osteoblastic differentiation compared with those on PCL or PCL-spherical nHA films. When cocultured with ASCs in an indirect coculture system, only the HOBs on PCL-rod nHA films up-regulated the gene expression of Runx2, bone sialoprotein, and osteocalcin of ASCs. Additionally, the HOBs on PCL-rod nHA films showed significant up-regulation of bone morphogenic protein 2 gene and protein expression and induced highest phosphorylated Smad1/5 protein level in ASCs. Treatment of the coculture medium with bone morphogenic protein 2 inhibitor (Noggin) largely abolished the osteogenic differentiation of the ASCs induced by the HOBs on PCL-rod nHA films. In conclusion, HOBs can not only best display their osteoblastic phenotype by culturing on PCL-rod nHA films but also render an optimal osteogenic niche for the differentiation of stem cells.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Durapatita/farmacología , Nanopartículas/química , Osteoblastos/citología , Osteogénesis/efectos de los fármacos , Poliésteres/farmacología , Células Madre/citología , Tejido Adiposo/citología , Proteína Morfogenética Ósea 2/antagonistas & inhibidores , Proteína Morfogenética Ósea 2/metabolismo , Adhesión Celular/efectos de los fármacos , Técnicas de Cocultivo , Humanos , Iones , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Nanopartículas/ultraestructura , Osteoblastos/efectos de los fármacos , Osteoblastos/ultraestructura , Transducción de Señal/efectos de los fármacos , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Nicho de Células Madre/citología , Nicho de Células Madre/efectos de los fármacos , Nicho de Células Madre/metabolismo , Propiedades de Superficie/efectos de los fármacos
16.
Dev Dyn ; 240(3): 737-43, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21337471

RESUMEN

A stem cell niche is a microenvironment that supports self-renewal of a population of stem cells, and their production of differentiated cells. While the definition evokes images of a stem cell Shangri-La-where a serene stem cell pool nestles within a niche that shelters and sustains it-the reality is much more tumultuous. Niches are subject to an ever-changing maelstrom of environmental factors, the ravages of old age, and the sly tactics of disease. Presented here is a basic overview of the different ways in which stem cell niches respond to local and systemic environments, and their impact on stem cell behavior. The primer culminates with a discussion of the topic with stem cell and niche biologists D. Leanne Jones, Ph.D., and Tudorita Tumbar, Ph.D.


Asunto(s)
Nicho de Células Madre/citología , Animales , Humanos , Entrevistas como Asunto , Nicho de Células Madre/metabolismo , Células Madre/citología , Células Madre/metabolismo
17.
Development ; 138(7): 1259-67, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21350008

RESUMEN

Interactions between niche cells and stem cells are vital for proper control over stem cell self-renewal and differentiation. However, there are few tissues where the initial establishment of a niche has been studied. The Drosophila testis houses two stem cell populations, which each lie adjacent to somatic niche cells. Although these niche cells sustain spermatogenesis throughout life, it is not understood how their fate is established. Here, we show that Notch signaling is necessary to specify niche cell fate in the developing gonad. Surprisingly, our results indicate that adjacent endoderm is the source of the Notch-activating ligand Delta. We also find that niche cell specification occurs earlier than anticipated, well before the expression of extant markers for niche cell fate. This work further suggests that endoderm plays a dual role in germline development. The endoderm assists both in delivering germ cells to the somatic gonadal mesoderm, and in specifying the niche where these cells will subsequently develop as stem cells. Because in mammals primordial germ cells also track through endoderm on their way to the genital ridge, our work raises the possibility that conserved mechanisms are employed to regulate germline niche formation.


Asunto(s)
Proteínas de Drosophila/metabolismo , Endodermo/metabolismo , Células Germinativas/metabolismo , Proteínas de la Membrana/metabolismo , Mesodermo/metabolismo , Receptores Notch/metabolismo , Nicho de Células Madre/metabolismo , Testículo/embriología , Animales , Animales Modificados Genéticamente , Drosophila/citología , Drosophila/embriología , Drosophila/metabolismo , Endodermo/citología , Células Germinativas/citología , Inmunohistoquímica , Hibridación in Situ , Péptidos y Proteínas de Señalización Intracelular , Masculino , Mesodermo/citología , Transducción de Señal/fisiología , Espermatogénesis/fisiología , Células Madre/citología , Células Madre/metabolismo , Testículo/citología , Testículo/metabolismo
18.
Cell ; 144(1): 92-105, 2011 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-21215372

RESUMEN

Here, we exploit the hair follicle to define the point at which stem cells (SCs) become irreversibly committed along a differentiation lineage. Employing histone and nucleotide double-pulse-chase and lineage tracing, we show that the early SC descendents en route to becoming transit-amplifying cells retain stemness and slow-cycling properties and home back to the bulge niche when hair growth stops. These become the primary SCs for the next hair cycle, whereas initial bulge SCs become reserves for injury. Proliferating descendents further en route irreversibly lose their stemness, although they retain many SC markers and survive, unlike their transit-amplifying progeny. Remarkably, these progeny also home back to the bulge. Combining purification and gene expression analysis with differential ablation and functional experiments, we define critical functions for these non-SC niche residents and unveil the intriguing concept that an irreversibly committed cell in an SC lineage can become an essential contributor to the niche microenvironment.


Asunto(s)
Folículo Piloso/citología , Folículo Piloso/crecimiento & desarrollo , Nicho de Células Madre/metabolismo , Células Madre/metabolismo , Animales , Antígenos CD34/metabolismo , Diferenciación Celular , Folículo Piloso/metabolismo , Humanos , Ratones , Piel/citología
19.
Blood ; 117(14): 3893-902, 2011 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-21224474

RESUMEN

Angiogenesis alleviates hypoxic stress in ischemic tissues or during tumor progression. In addition to endothelial cell proliferation and migration, the angiogenic process requires bone marrow-derived cell (BMDC) recruitment to sites of neovascularization. However, the mechanism of communication between hypoxic tissues and the BM remains unknown. Using 2 models of hypoxia-induced angiogenesis (ischemic hindlimb surgery and subcutaneous tumor growth), we show that platelet infusion promotes BMDC mobilization into the circulation, BMDC recruitment into growing neovasculature, tumor vascularization, and blood flow restoration in ischemic limbs, whereas platelet depletion inhibits these effects. Thus, platelets are required for BMDC recruitment into ischemia-induced vasculature. Secretion of platelet α-granules, but neither dense granules nor platelet aggregation is crucial for BMDC homing and subsequent angiogenesis, as determined using VAMP-8(-/-), Pearl, and integrin Beta 3(-/-) platelets. Finally, platelets sequester tumor-derived promoters of angiogenesis and BMDC mobilization, which are counterbalanced by the antiangiogenic factor thrombospondin-1. A lack of thrombospondin-1 in platelets leads to an imbalance in proangiogenic and antiangiogenic factors and accelerates tumor growth and vascularization. Our data demonstrate that platelets stimulate BMDC homing in a VAMP-8-dependent manner, revealing a previously unknown role for platelets as key mediators between hypoxic tissues and the bone marrow during angiogenesis.


Asunto(s)
Plaquetas/metabolismo , Plaquetas/fisiología , Células de la Médula Ósea/fisiología , Movimiento Celular , Neovascularización Patológica/etiología , Animales , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Procesos de Crecimiento Celular/genética , Movimiento Celular/fisiología , Femenino , Hipoxia/patología , Hipoxia/fisiopatología , Integrina beta3/genética , Integrina beta3/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Neoplasias/irrigación sanguínea , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Estrés Oxidativo/fisiología , Proteínas R-SNARE/genética , Proteínas R-SNARE/fisiología , Nicho de Células Madre/metabolismo , Nicho de Células Madre/patología , Trombospondina 1/genética , Trombospondina 1/fisiología , Células Tumorales Cultivadas
20.
Acta Pharmacol Sin ; 32(2): 152-9, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21217770

RESUMEN

Embryonic stem (ES) cells and their differentiated progeny offer tremendous potential for regenerative medicine, even in the field of drug discovery. There is an urgent need for clinically relevant assays that make use of ES cells because of their rich biological utility. Attention has been focused on small molecules that allow the precise manipulation of cells in vitro, which could allow researchers to obtain homogeneous cell types for cell-based therapies and discover drugs for stimulating the regeneration of endogenous cells. Such therapeutics can act on target cells or their niches in vivo to promote cell survival, proliferation, differentiation, and homing. In the present paper, we reviewed the use of ES cell models for high-throughput/content drug screening and toxicity assessment. In addition, we examined the role of stem cells in large pharmaceutical companies' R&D and discussed a novel subject, nicheology, in stem cell-related research fields.


Asunto(s)
Descubrimiento de Drogas/métodos , Células Madre Embrionarias/metabolismo , Medicina Regenerativa/métodos , Animales , Sistemas de Liberación de Medicamentos , Industria Farmacéutica/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Modelos Biológicos , Nicho de Células Madre/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...