Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 217
Filtrar
1.
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
2.
Development ; 138(16): 3367-76, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21752931

RESUMEN

Adult stem cells modulate their output by varying between symmetric and asymmetric divisions, but have rarely been observed in living intact tissues. Germline stem cells (GSCs) in the Drosophila testis are anchored to somatic hub cells and were thought to exclusively undergo oriented asymmetric divisions, producing one stem cell that remains hub-anchored and one daughter cell displaced out of the stem cell-maintaining micro-environment (niche). We developed extended live imaging of the Drosophila testis niche, allowing us to track individual germline cells. Surprisingly, new wild-type GSCs are generated in the niche during steady-state tissue maintenance by a previously undetected event we term 'symmetric renewal', where interconnected GSC-daughter cell pairs swivel such that both cells contact the hub. We also captured GSCs undergoing direct differentiation by detaching from the hub. Following starvation-induced GSC loss, GSC numbers are restored by symmetric renewals. Furthermore, upon more severe (genetically induced) GSC loss, both symmetric renewal and de-differentiation (where interconnected spermatogonia fragment into pairs while moving towards then establishing contact with the hub) occur simultaneously to replenish the GSC pool. Thus, stereotypically oriented stem cell divisions are not always correlated with an asymmetric outcome in cell fate, and changes in stem cell output are governed by altered signals in response to tissue requirements.


Asunto(s)
Drosophila melanogaster/citología , Microscopía Confocal/métodos , Espermatogonias/citología , Nicho de Células Madre/citología , Células Madre/citología , Animales , Diferenciación Celular , Linaje de la Célula , Supervivencia Celular , Masculino , Testículo/citología
3.
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
5.
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
6.
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
7.
Blood ; 118(6): 1516-24, 2011 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-21673348

RESUMEN

A large body of evidence suggests hemopoietic stem cells (HSCs) exist in an endosteal niche close to bone, whereas others suggest that the HSC niche is intimately associated with vasculature. In this study, we show that transplanted hemopoietic stem and progenitor cells (HSPCs) home preferentially to the trabecular-rich metaphysis of the femurs in nonablated mice at all time points from 15 minutes to 15 hours after transplantation. Within this region, they exist in an endosteal niche in close association with blood vessels. The preferential homing of HSPCs to the metaphysis occurs rapidly after transplantation, suggesting that blood vessels within this region may express a unique repertoire of endothelial adhesive molecules. One candidate is hyaluronan (HA), which is highly expressed on the blood vessel endothelium in the metaphysis. Analysis of the early stages of homing and the spatial dis-tribution of transplanted HSPCs at the single-cell level in mice devoid of Has3-synthesized HA, provides evidence for a previously undescribed role for HA expressed on endothelial cells in directing the homing of HSPCs to the metaphysis.


Asunto(s)
Vasos Sanguíneos/citología , Médula Ósea/irrigación sanguínea , Huesos/citología , Células Madre Hematopoyéticas/citología , Animales , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/ultraestructura , Huesos/metabolismo , Moléculas de Adhesión Celular/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Femenino , Fémur/citología , Fémur/metabolismo , Glucuronosiltransferasa/genética , Glucuronosiltransferasa/metabolismo , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Hialuronano Sintasas , Ácido Hialurónico/metabolismo , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Nicho de Células Madre/irrigación sanguínea , Nicho de Células Madre/citología , Migración Transendotelial y Transepitelial , Microtomografía por Rayos X
8.
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
9.
Eur J Dermatol ; 21 Suppl 2: 21-8, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21628126

RESUMEN

Transition zones (TZs) are regions in the body where two different types of epithelial tissue meet resulting in the appearance of a distinct abrupt transition. These TZs are found in numerous locations within the body, including the cornea-conjunctiva junction, esophagogastric junction, gastro-duodenal junction, endo-ectocervix junction, ileocecal junction, and anorectal junction. Several of these TZs are often associated with the development of cancer, in some cases due to viral transformation by the human papilloma virus (HPV). The underlying molecular and cellular basis for this tumor susceptibiblity is unknown. The distinct epithelial morphology and location results in unique properties being conferred upon this epithelial tissue, as different signaling cues and cell surface markers are apparent. Importantly, the natural state of TZs closely resembles that of a pre-lesional epithelium, as several proteins that are induced during wounding are expressed specifically within this region, which may contribute to transformation. This region may also act as a stem cell niche, and as such, represents a key location for cellular transformation by accumulated genetic mutations or viral transformation resulting in tumor formation.


Asunto(s)
Epitelio/fisiología , Nicho de Células Madre/citología , Células Madre/fisiología , Carcinoma de Células Escamosas/patología , Transformación Celular Neoplásica/patología , Cuello del Útero/patología , Células Epiteliales/fisiología , Unión Esofagogástrica/citología , Unión Esofagogástrica/fisiología , Femenino , Humanos , Inmunomodulación/fisiología , Queratina-17/fisiología , Queratinocitos , Limbo de la Córnea/citología , Papillomaviridae/genética
10.
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
11.
BMC Neurosci ; 12: 53, 2011 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-21635768

RESUMEN

BACKGROUND: Adult neurogenesis, the production and integration of new neurons into circuits in the brains of adult animals, is a common feature of a variety of organisms, ranging from insects and crustaceans to birds and mammals. In the mammalian brain the 1st-generation neuronal precursors, the astrocytic stem cells, reside in neurogenic niches and are reported to undergo self-renewing divisions, thereby providing a source of new neurons throughout an animal's life. In contrast, our work shows that the 1st-generation neuronal precursors in the crayfish (Procambarus clarkii) brain, which also have glial properties and lie in a neurogenic niche resembling that of vertebrates, undergo geometrically symmetrical divisions and both daughters appear to migrate away from the niche. However, in spite of this continuous efflux of cells, the number of neuronal precursors in the crayfish niche continues to expand as the animals grow and age. Based on these observations we have hypothesized that (1) the neuronal stem cells in the crayfish brain are not self-renewing, and (2) a source external to the neurogenic niche must provide cells that replenish the stem cell pool. RESULTS: In the present study, we tested the first hypothesis using sequential double nucleoside labeling to track the fate of 1st- and 2nd-generation neuronal precursors, as well as testing the size of the labeled stem cell pool following increasing incubation times in 5-bromo-2'-deoxyuridine (BrdU). Our results indicate that the 1st-generation precursor cells in the crayfish brain, which are functionally analogous to neural stem cells in vertebrates, are not a self-renewing population. In addition, these studies establish the cycle time of these cells. In vitro studies examining the second hypothesis show that Cell Tracker™ Green-labeled cells extracted from the hemolymph, but not other tissues, are attracted to and incorporated into the neurogenic niche, a phenomenon that appears to involve serotonergic mechanisms. CONCLUSIONS: These results challenge our current understanding of self-renewal capacity as a defining characteristic of all adult neuronal stem cells. In addition, we suggest that in crayfish, the hematopoietic system may be a source of cells that replenish the niche stem cell pool.


Asunto(s)
Células Madre Adultas/citología , Encéfalo/citología , Células-Madre Neurales/citología , Neurogénesis/fisiología , Neuronas/citología , Nicho de Células Madre/citología , Células Madre Adultas/fisiología , Animales , Astacoidea , Astrocitos/citología , Astrocitos/fisiología , Encéfalo/fisiología , Glutamato-Amoníaco Ligasa/metabolismo , Células-Madre Neurales/fisiología , Neuronas/fisiología , Serotonina/metabolismo , Nicho de Células Madre/fisiología
12.
Nature ; 474(7350): 216-9, 2011 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-21654805

RESUMEN

Stem cells reside in a specialized regulatory microenvironment or niche, where they receive appropriate support for maintaining self-renewal and multi-lineage differentiation capacity. The niche may also protect stem cells from environmental insults including cytotoxic chemotherapy and perhaps pathogenic immunity. The testis, hair follicle and placenta are all sites of residence for stem cells and are immune-suppressive environments, called immune-privileged sites, where multiple mechanisms cooperate to prevent immune attack, even enabling prolonged survival of foreign allografts without immunosuppression. We sought to determine if somatic stem-cell niches more broadly are immune-privileged sites by examining the haematopoietic stem/progenitor cell (HSPC) niche in the bone marrow, a site where immune reactivity exists. We observed persistence of HSPCs from allogeneic donor mice (allo-HSPCs) in non-irradiated recipient mice for 30 days without immunosuppression with the same survival frequency compared to syngeneic HSPCs. These HSPCs were lost after the depletion of FoxP3 regulatory T (T(reg)) cells. High-resolution in vivo imaging over time demonstrated marked co-localization of HSPCs with T(reg) cells that accumulated on the endosteal surface in the calvarial and trabecular bone marrow. T(reg) cells seem to participate in creating a localized zone where HSPCs reside and where T(reg) cells are necessary for allo-HSPC persistence. In addition to processes supporting stem-cell function, the niche will provide a relative sanctuary from immune attack.


Asunto(s)
Supervivencia de Injerto/inmunología , Células Madre Hematopoyéticas/inmunología , Imagenología Tridimensional , Nicho de Células Madre/inmunología , Linfocitos T Reguladores/inmunología , Animales , Supervivencia Celular/inmunología , Células Cultivadas , Factores de Transcripción Forkhead/metabolismo , Células Madre Hematopoyéticas/citología , Humanos , Interleucina-10/deficiencia , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-10/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Nicho de Células Madre/citología , Linfocitos T Reguladores/metabolismo , Factores de Tiempo , Trasplante Homólogo/inmunología
13.
Endocrinology ; 152(8): 2957-62, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21610157

RESUMEN

Long known to be home to hematopoietic stem cells (HSC), the bone/bone marrow organ and its cellular components are directly implicated in regulating hematopoiesis and HSC function. Over the past few years, advances on the identity of HSC "niche" cells have brought into focus the role of cells of osteogenic lineage and of marrow microvessels. At the same time, the identity of self-renewing multipotent skeletal progenitors (skeletal stem cells, also known as mesenchymal stem cells) has also been more precisely defined, along with the recognition of their own microvascular niche. The two sets of evidence converge in delineating a picture in which two kinds of stem cells share an identical microanatomical location in the bone/bone marrow organ. This opens a new view on the manner in which the skeleton and hematopoiesis can cross-regulate via interacting stem cells but also a novel view of our general concept of stem cell niches.


Asunto(s)
Hematopoyesis , Células Madre Hematopoyéticas/citología , Células Madre Mesenquimatosas/citología , Osteogénesis , Nicho de Células Madre/citología , Animales , Humanos , Morfogénesis
14.
Methods Mol Biol ; 750: 117-29, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21618087

RESUMEN

A stem cell niche is a specialized tissue environment that controls the proliferation and differentiation of its resident stem cells. The functions of these structures have been well characterized in adult organisms. In particular, the bone marrow stem cell niche in mammals has been amenable to analysis because of the ability of transplanted hematopoietic cells to home and to recolonize the bone marrow of an irradiated host. Despite clues from adult models, it remains unclear how stem cells become partitioned into appropriate niches during embryonic development. To examine the earliest steps in niche formation, we created an organ culture system to observe the development of primordial germ cells (PGCs), a migratory stem cell population that will eventually give rise to the gametes. Using this assay, we can watch PGCs as they migrate to colonize the developing gonads and can introduce growth factor agonists or antagonists to test the function of proteins that regulate this process. This provides an unprecedented opportunity to identify the cellular and molecular interactions required for the formation of the germ cell niche.


Asunto(s)
Movimiento Celular , Embrión de Mamíferos/citología , Células Madre Embrionarias/citología , Células Germinativas , Técnicas de Cultivo de Órganos/métodos , Nicho de Células Madre/citología , Animales , Diferenciación Celular , Supervivencia Celular , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/metabolismo , Femenino , Fluorescencia , Células Germinativas/citología , Células Germinativas/metabolismo , Proteínas Fluorescentes Verdes/análisis , Masculino , Ratones , Ratones Transgénicos , Microscopía Confocal , Modelos Animales , Embarazo , Nicho de Células Madre/embriología , Imagen de Lapso de Tiempo
15.
Methods Mol Biol ; 750: 197-214, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21618093

RESUMEN

The tracking of immunofluorescent labeled hematopoietic stem and progenitor cells (HSC/HPC) within the bone marrow (BM) cavity allows the assessment of the regulatory processes involved in transendothelial migration, trans-marrow migration, and finally lodgement into the HSC niche. This is of interest as the extracellular and cellular components involved in the regulation of HSC quiescence and differentiation are still not completely understood. Homing of transplanted HSC is the first critical step in the interaction between HSC and the microenvironment of the BM. As a consequence, murine models allowing the evaluation of the structural relationship between migrating HSC, the endosteal bone surface, and the vascular components of the BM enhance our understanding of hematopoietic regulation.


Asunto(s)
Médula Ósea/fisiología , Movimiento Celular , Endotelio Vascular/citología , Células Madre Hematopoyéticas/citología , Nicho de Células Madre/citología , Migración Transendotelial y Transepitelial/fisiología , Animales , Benzopiranos/análisis , Diferenciación Celular , Endotelio Vascular/fisiología , Citometría de Flujo , Fluoresceínas/análisis , Colorantes Fluorescentes/análisis , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/fisiología , Separación Inmunomagnética , Ratones , Ratones Endogámicos C57BL , Naftoles/análisis , Rodaminas/análisis , Nicho de Células Madre/fisiología , Succinimidas/análisis
16.
Methods Mol Biol ; 750: 215-24, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21618094

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs) are located in the bone marrow in zones of residence specialized in supporting them which are referred to as niches. It is in such a specialized niche that normal HSPCs are maintained to perform their self-renewal and differentiation duties in a highly controlled manner. One challenge in dissecting the functional significance of the complex cellular and molecular interactions in the niche is to link the types and qualities of cell-cell contacts to the intracellular signaling components involved in cell regulation. Attempts to study the interactions of HSPC with their niche eventually have to be performed in their natural location in vivo, as isolation of the cells from bone -marrow will disrupt the HSPC-niche interactions and thus not reveal functionally critical cell-cell -contacts. Intravital imaging of individual cells in the bone marrow has just recently been introduced, almost exclusively focusing on imaging inside the marrow of the calvaria. However, calvarial marrow is functionally distinct from marrow of long bones, the major source of HSPC for both physiology and study. To overcome these limitations, we developed a novel method for multiphoton intravital imaging of HSPC in the marrow of long bones.


Asunto(s)
Médula Ósea/fisiología , Células Madre Hematopoyéticas/citología , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Imagen Molecular/métodos , Nicho de Células Madre/citología , Tibia/citología , Animales , Comunicación Celular/fisiología , Diferenciación Celular , Movimiento Celular , Células Madre Hematopoyéticas/fisiología , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Nicho de Células Madre/fisiología
17.
Methods Mol Biol ; 750: 261-74, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21618097

RESUMEN

It is likely that adult epithelial stem cells will be useful in the treatment of diseases, such as ectodermal dysplasias, monilethrix, Netherton syndrome, Menkes disease, hereditary epidermolysis bullosa, and alopecias. Additionally, other skin problems such as burn wounds, chronic wounds, and ulcers will benefit from stem cell-related therapies. However, there are many questions that need to be answered before this goal can be realized. The most important of these questions is what regulates the adhesion of stem cells to the niche versus migration to the site of injury. We have started to identify the mechanisms involved in this decision-making process.


Asunto(s)
Células Madre Adultas/citología , Diferenciación Celular/fisiología , Movimiento Celular , Células Epiteliales/citología , Folículo Piloso/citología , Queratinocitos/citología , Nicho de Células Madre/citología , Adulto , Células Madre Adultas/fisiología , Células Madre Adultas/trasplante , Animales , Adhesión Celular , Células Epiteliales/fisiología , Células Epiteliales/trasplante , Citometría de Flujo , Folículo Piloso/fisiología , Humanos , Inmunohistoquímica , Queratinocitos/fisiología , Ratones , Enfermedades de la Piel/terapia , Nicho de Células Madre/fisiología , Trasplante de Células Madre/métodos , Imagen de Lapso de Tiempo
18.
Regen Med ; 6(3): 351-66, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21548740

RESUMEN

Osteoarthritis is a common disorder in which there is not only extensive degeneration but also an aberrant attempt at repair in joints. Stem cell therapy could provide a permanent, biological solution, with all sources of stem cells (embryonic, fetal and adult) showing some degree of potential. Mesenchymal stromal/stem cells, however, appear to be the leading candidates because of their ability to be sourced from many or all joint tissues. They may also modulate the immune response of individuals, in a manner influenced by local factors. This biological behavior of stem cells renders the application of regulatory standardizations challenging in comparison to pharmaceutical therapies. However, this would not be an issue if endogenous stem cells were activated to effect repair of an arthritic joint.


Asunto(s)
Osteoartritis/terapia , Trasplante de Células Madre , Humanos , Articulaciones/patología , Osteoartritis/patología , Nicho de Células Madre/citología , Células Madre/citología
19.
Curr Opin Hematol ; 18(4): 214-9, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21537167

RESUMEN

PURPOSE OF REVIEW: The maintenance of a functional hematopoietic niche is critical for modulating the fate of hematopoietic stem cells (HSCs). Several enzymes were described as essential for guaranteeing niche functionality. This review summarizes the recent findings about the role of galactocerebrosidase and other enzymes involved in the maintenance of a functional HSC niche. RECENT FINDINGS: The essential role of enzymes actively involved in the maintenance of the bone marrow microenvironment, in bone remodeling, in regulating the sympathetic innervation of the niche, and in the production and relative balance of sphingolipids active in the niche has been recently highlighted. Enzymes involved in bone remodeling modify the cell-to-cell interaction between osteoblasts and HSCs. Heparanase, neutrophil elastase, and alpha-iduronidase affect the bioavailability of key cytokines and ligands within the extracellular matrix of the niche. Moreover, galactosyltransferase and galactocerebrosidase affect the function of the sympathetic nervous system and/or the balance of bioactive sphingolipids, thus influencing the SDF-1/CXCR4 axis and the proliferation of HSCs. SUMMARY: Here, we discuss the role of different enzymes directly or indirectly influencing the niche microenvironment, and we provide a comprehensive picture of their cooperative role, together with receptors, soluble factors, and the extracellular matrix, in maintaining a functional hematopoietic niche.


Asunto(s)
Galactosilceramidasa/metabolismo , Células Madre Hematopoyéticas/enzimología , Nicho de Células Madre/enzimología , Animales , Células Madre Hematopoyéticas/citología , Humanos , Ratones , Nicho de Células Madre/citología
20.
Curr Opin Hematol ; 18(4): 231-8, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21537168

RESUMEN

PURPOSE OF REVIEW: Blood cell production is maintained by hematopoietic stem cells (HSCs) that reside in specialized niches within bone marrow. Treatment with granulocyte-colony stimulating factor (G-CSF) causes HSC egress from bone marrow niches and trafficking to the peripheral blood, a process termed 'mobilization'. Although the mobilization phenomenon has been known for some time and is utilized clinically to acquire HSC for transplant, the mechanisms mediating HSC release are not completely understood. We discuss recent advances and controversies in defining the mechanisms responsible for G-CSF-induced mobilization. RECENT FINDINGS: New reports define a role for resident monocytes/macrophages in maintaining niche cells, which is diminished after G-CSF treatment, suggesting a new mechanism for mobilization. Although osteoblasts have been reported to be a primary component of the HSC niche, new results suggest a unique niche composed of innervated mesenchymal stem cells. Modulating bioactive lipid signaling also facilitates mobilization, and may define a future therapeutic strategy. SUMMARY: Hematopoietic mobilization by G-CSF is primarily mediated by alterations to the bone marrow niche by both direct and indirect mechanisms, rather than directly altering HSC function. Further understanding of the processes mediating mobilization will advance our understanding on the cellular and molecular components of the HSC niche.


Asunto(s)
Movimiento Celular/fisiología , Células Madre Hematopoyéticas/citología , Nicho de Células Madre/citología , Animales , Movimiento Celular/efectos de los fármacos , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Ratones , Nicho de Células Madre/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...