Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Cutan Pathol ; 51(7): 490-495, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38548658

RESUMEN

Dermatofibrosarcoma protuberans (DFSP) is a cutaneous sarcoma with a high propensity for local invasion and recurrence. Although it is a rare event, the occurrence of multiple tumors in a single patient raises a diagnostic dilemma, as metastatic disease should be differentiated from multiple primary malignant events. In more than 90% of DFSP, a pathogenic t(17;22) translocation leads to the expression of COL1A1::PDGFB fusion transcripts. Karyotype analysis, fluorescence in situ hybridization, and RT-PCR can be useful ancillary studies in detecting this characteristic rearrangement, and sequencing of the fusion transcript can be used to support a clonal origin in metastatic and multifocal disease. However, previous reports have demonstrated variable sensitivity of these assays, in part due to the high sequence variability of the COL1A1::PDGFB fusion. Here, we report a patient who developed two distinct DFSP tumors over the course of 7 years. Chromosomal microarray analysis identified distinctive genomic alterations in the two tumors, supporting the occurrence of multiple primary malignant events.


Asunto(s)
Dermatofibrosarcoma , Proteínas de Fusión Oncogénica , Neoplasias Cutáneas , Humanos , Masculino , Cromosomas Humanos Par 17/genética , Cromosomas Humanos Par 22/genética , Cadena alfa 1 del Colágeno Tipo I , Dermatofibrosarcoma/genética , Dermatofibrosarcoma/patología , Dermatofibrosarcoma/diagnóstico , Hibridación Fluorescente in Situ/métodos , Análisis por Micromatrices/métodos , Neoplasias Primarias Múltiples/genética , Neoplasias Primarias Múltiples/patología , Proteínas de Fusión Oncogénica/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Translocación Genética , Persona de Mediana Edad
2.
Allergy Asthma Proc ; 40(5): 350-353, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31514794

RESUMEN

Angioedema is a potentially life-threatening swelling condition that can occur either in isolation or in the context of other syndromes, e.g., anaphylaxis. Angioedema is typically asymmetric, lasts for hours to days, is not gravity dependent, and is often nonpitting. Recurrent angioedema is typically associated with histaminergic and bradykinin-mediated causes, some of which can indicate underlying etiologies with high morbidity or mortality. The differential diagnosis for acute angioedema can include anaphylaxis, chronic urticaria with angioedema, medications such as angiotensin-converting-enzyme inhibitors, hereditary C1 esterase inhibitor defects, and acquired defects; however, the cause is often idiopathic, and effective therapy can be elusive. In this article, we described a unique etiology of a case of isolated recurrent angioedema that improved when the possible underlying cause was successfully treated.


Asunto(s)
Angioedema/etiología , Anciano de 80 o más Años , Angioedema/diagnóstico , Diagnóstico Diferencial , Humanos , Masculino , Recurrencia
4.
Front Oncol ; 14: 1413793, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39136002

RESUMEN

Merkel cell carcinoma (MCC) is an aggressive cutaneous neuroendocrine carcinoma thought to arise via either viral (Merkel cell polyomavirus) or ultraviolet-associated pathways. Surgery and radiotherapy have historically been mainstays of management, and immunotherapy has improved outcomes for advanced disease. However, there remains a lack of effective therapy for those patients who fail to respond to these established approaches, underscoring a critical need to better understand MCC biology for more effective prognosis and treatment. Here, we review the fundamental aspects of MCC biology and the recent advances which have had profound impact on management. The first genetically-engineered mouse models for MCC tumorigenesis provide opportunities to understand the potential MCC cell of origin and may prove useful for preclinical investigation of novel therapeutics. The MCC cell of origin debate has also been advanced by recent observations of MCC arising in association with a clonally related hair follicle tumor or squamous cell carcinoma in situ. These studies also suggested a role for epigenetics in the origin of MCC, highlighting a potential utility for this therapeutic avenue in MCC. These and other therapeutic targets form the basis for a wealth of ongoing clinical trials to improve MCC management. Here, we review these recent advances in the context of the existing literature and implications for future investigations.

5.
JCI Insight ; 5(13)2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32544094

RESUMEN

Wnt/ß-catenin signaling is active in small subpopulations of Ewing sarcoma cells, and these cells display a more metastatic phenotype, in part due to antagonism of EWS-FLI1-dependent transcriptional activity. Importantly, these ß-catenin-activated Ewing sarcoma cells also alter secretion of extracellular matrix (ECM) proteins. We thus hypothesized that, in addition to cell-autonomous mechanisms, Wnt/ß-catenin-active tumor cells might contribute to disease progression by altering the tumor microenvironment (TME). Analysis of transcriptomic data from primary patient biopsies and from ß-catenin-active versus -nonactive tumor cells identified angiogenic switch genes as being highly and reproducibly upregulated in the context of ß-catenin activation. In addition, in silico and in vitro analyses, along with chorioallantoic membrane assays, demonstrated that ß-catenin-activated Ewing cells secreted factors that promote angiogenesis. In particular, activation of canonical Wnt signaling leads Ewing sarcoma cells to upregulate expression and secretion of proangiogenic ECM proteins, collectively termed the angiomatrix. Significantly, our data show that induction of the angiomatrix by Wnt-responsive tumor cells is indirect and is mediated by TGF-ß. Mechanistically, Wnt/ß-catenin signaling antagonizes EWS-FLI1-dependent repression of TGF-ß receptor type 2, thereby sensitizing tumor cells to TGF-ß ligands. Together, these findings suggest that Wnt/ß-catenin-active tumor cells can contribute to Ewing sarcoma progression by promoting angiogenesis in the local TME.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Sarcoma de Ewing/metabolismo , Microambiente Tumoral/fisiología , Vía de Señalización Wnt/fisiología , Línea Celular Tumoral , Humanos , Proteínas de Fusión Oncogénica/genética , Proteína Proto-Oncogénica c-fli-1/genética , Proteína EWS de Unión a ARN/genética , Sarcoma de Ewing/genética , Sarcoma de Ewing/patología , Regulación hacia Arriba
6.
J Cell Biochem ; 105(2): 370-80, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18636554

RESUMEN

One of the most life-threatening complications of prostate cancer is skeletal metastasis. In order to develop treatment for metastasis, it is important to understand its molecular mechanisms. Our work in this field has drawn parallels between hematopoietic stem cell and prostate cancer homing to the marrow. Our recent work demonstrated that annexin II expressed by osteoblasts and endothelial cells plays a critical role in niche selection. In this study, we demonstrate that annexin II and its receptor play a crucial role in establishing metastasis of prostate cancer. Prostate cancer cell lines migrate toward annexin II and the adhesion of prostate cancer to osteoblasts and endothelial cells was inhibited by annexin II. By blocking annexin II or its receptor in animal models, short-term and long-term localization of prostate cancers are limited. Annexin II may also facilitate the growth of prostate cancer in vitro and in vivo by the MAPK pathway. These data strongly suggest that annexin II and its receptor axis plays a central role in prostate cancer metastasis, and that prostate cancer utilize the hematopoietic stem cell homing mechanisms to gain access to the niche.


Asunto(s)
Anexina A2/fisiología , Fenómenos Fisiológicos Celulares , Metástasis de la Neoplasia/patología , Neoplasias de la Próstata/patología , Receptores de Péptidos/fisiología , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Técnicas de Cocultivo , Células Endoteliales , Humanos , Masculino , Osteoblastos
7.
Clin Exp Metastasis ; 25(7): 765-76, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18563594

RESUMEN

Stromal derived factor-1 (SDF-1 or CXCL12) expressed by osteoblasts and endothelial cells, and its receptors CXCR4 and CXCR7/RDC1 are key molecular determinants in prostate cancer (PCa) metastasis. What drives PCa cells into the extravascular marrow space(s) once they make contact with the blood vessel endothelium, however remains unclear. Here, we evaluated whether degradation of CXCL12 facilitates PCa cell entry into the marrow cavity by locally lowering CXCL12 levels intravascularly. To explore this possibility, co-cultured conditioned media from PCa cells and endothelial cells were evaluated for their ability to degrade biotinylated CXCL12 (bCXCL12). Co-culture of PCa cells/endothelial cells resulted in greater digestion of CXCL12 than was achieved by either cell type alone, and this activity regulated invasion in vitro. The ability to degrade CXCL12 was not however observed in PCa and osteoblasts co-cultures. Fractionation and inhibitor studies suggested that the activity was CD26/dipeptidyl peptidase IV (DPPIV) and possibly other cysteine/serine proteases. By inhibiting CD26/DPPIV, invasion and metastasis of PCa cell lines were enhanced in in vitro and in vivo metastasis assays. Together, these data suggest that the degradation of CXCL12 by CD26/DPPIV may be involved in the metastatic cascades of PCa, and suggests that inhibition of CD26/DPPIV may be a trigger of PCa metastasis.


Asunto(s)
Quimiocina CXCL12/metabolismo , Dipeptidil Peptidasa 4/fisiología , Neoplasias de la Próstata/patología , Animales , Quimiocina CXCL12/análisis , Técnicas de Cocultivo , Cisteína Endopeptidasas/fisiología , Humanos , Masculino , Ratones , Ratones SCID , Metástasis de la Neoplasia , Serina Endopeptidasas/fisiología
12.
Neoplasia ; 18(8): 480-8, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27566104

RESUMEN

Metastatic Ewing sarcoma has a very poor prognosis and therefore new investigations into the biologic drivers of metastatic progression are key to finding new therapeutic approaches. The tumor microenvironment is highly dynamic, leading to exposure of different regions of a growing solid tumor to changes in oxygen and nutrient availability. Tumor cells must adapt to such stress in order to survive and propagate. In the current study, we investigate how Ewing sarcoma cells respond to the stress of growth factor deprivation and hypoxia. Our findings reveal that serum deprivation leads to a reversible change in Ewing cell cytoskeletal phenotypes. Using an array of migration and invasion techniques, including gelatin matrix degradation invadopodia assays, we show that exposure of Ewing sarcoma cells to serum deprivation and hypoxia triggers enhanced migration, invadopodia formation, matrix degradation and invasion. Further, these functional changes are accompanied by and dependent on activation of Src kinase. Activation of Src, and the associated invasive cell phenotype, were blocked by exposing hypoxia and serum-deprived cells to the Src inhibitor dasatinib. These results indicate that Ewing sarcoma cells demonstrate significant plasticity in response to rapidly changing micro-environmental stresses that can result from rapid tumor growth and from necrosis-causing therapies. In response to these stresses, Ewing cells transition to a more migratory and invasive state and our data show that Src is an important mediator of this stress response. Our data support exploration of clinically available Src inhibitors as adjuvant agents for metastasis prevention in Ewing sarcoma.


Asunto(s)
Podosomas/metabolismo , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patología , Estrés Fisiológico , Microambiente Tumoral , Familia-src Quinasas/metabolismo , Actinas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Dasatinib/farmacología , Activación Enzimática/efectos de los fármacos , Transición Epitelial-Mesenquimal , Matriz Extracelular/metabolismo , Humanos , Hipoxia/metabolismo , Fenotipo , Estrés Fisiológico/efectos de los fármacos
13.
Cancer Res ; 76(17): 5040-53, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27364557

RESUMEN

Ewing sarcomas are characterized by the presence of EWS/ETS fusion genes in the absence of other recurrent genetic alterations and mechanisms of tumor heterogeneity that contribute to disease progression remain unclear. Mutations in the Wnt/ß-catenin pathway are rare in Ewing sarcoma but the Wnt pathway modulator LGR5 is often highly expressed, suggesting a potential role for the axis in tumor pathogenesis. We evaluated ß-catenin and LGR5 expression in Ewing sarcoma cell lines and tumors and noted marked intra- and inter-tumor heterogeneity. Tumors with evidence of active Wnt/ß-catenin signaling were associated with increased incidence of tumor relapse and worse overall survival. Paradoxically, RNA sequencing revealed a marked antagonism of EWS/ETS transcriptional activity in Wnt/ß-catenin-activated tumor cells. Consistent with this, Wnt/ß-catenin-activated cells displayed a phenotype that was reminiscent of Ewing sarcoma cells with partial EWS/ETS loss of function. Specifically, activation of Wnt/ß-catenin induced alterations to the actin cytoskeleton, acquisition of a migratory phenotype, and upregulation of EWS/ETS-repressed genes. Notably, activation of Wnt/ß-catenin signaling led to marked induction of tenascin C (TNC), an established promoter of cancer metastasis, and an EWS/ETS-repressed target gene. Loss of TNC function in Ewing sarcoma cells profoundly inhibited their migratory and metastatic potential. Our studies reveal that heterogeneous activation of Wnt/ß-catenin signaling in subpopulations of tumor cells contributes to phenotypic heterogeneity and disease progression in Ewing sarcoma. Significantly, this is mediated, at least in part, by inhibition of EWS/ETS fusion protein function that results in derepression of metastasis-associated gene programs. Cancer Res; 76(17); 5040-53. ©2016 AACR.


Asunto(s)
Neoplasias Óseas/patología , Sarcoma de Ewing/patología , Vía de Señalización Wnt/fisiología , Animales , Western Blotting , Neoplasias Óseas/metabolismo , Proteínas de Unión a Calmodulina/metabolismo , Línea Celular Tumoral , Citometría de Flujo , Xenoinjertos , Humanos , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas de Fusión Oncogénica/metabolismo , Fenotipo , Reacción en Cadena de la Polimerasa , Proteína Proto-Oncogénica c-ets-1/metabolismo , Proteína EWS de Unión a ARN , Proteínas de Unión al ARN/metabolismo , Sarcoma de Ewing/metabolismo , Análisis de Matrices Tisulares , Transcriptoma
14.
Front Oncol ; 3: 81, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23596566

RESUMEN

Ewing sarcoma (ES) is an aggressive bone and soft tissue tumor of putative stem cell origin that predominantly occurs in children and young adults. Although most patients with localized ES can be cured with intensive therapy, the clinical course is variable and up to one third of patients relapse following initial remission. Unfortunately, little is yet known about the biologic features that distinguish low-risk from high-risk disease or the mechanisms of ES disease progression. Recent reports have suggested that putative cancer stem cells exist in ES and may contribute to an aggressive phenotype. The cell surface receptor leucine-rich repeat-containing G-protein coupled receptor 5 (LGR5) is a somatic stem cell marker that functions as an oncogene in several human cancers, most notably colorectal carcinoma. LGR5 is a receptor for the R-spondin (RSPO) family of ligands and RSPO-mediated activation of LGR5 potentiates Wnt/ß-catenin signaling, contributing to stem cell proliferation and self-renewal. Given its presumed stem cell origin, we investigated whether LGR5 contributes to ES pathogenesis. We found that LGR5 is expressed by ES and that its expression is relatively increased in cells and tumors that display a more aggressive phenotype. In particular, LGR5 expression was increased in putative cancer stem cells. We also found that neural crest-derived stem cells express LGR5, raising the possibility that expression of LGR5 may be a feature of ES cells of origin. LGR5-high ES cells showed nuclear localization of ß-catenin and robust activation of TCF reporter activity when exposed to Wnt ligand and this was potentiated by RSPO. However, modulation of LGR5 or exposure to RSPO had no impact on proliferation confirming that Wnt/ß-catenin signaling in ES cells does not recapitulate signaling in epithelial cells. Together these studies show that the RSPO-LGR5-Wnt-ß-catenin axis is present and active in ES and may contribute to tumor pathogenesis.

15.
Front Biosci (Schol Ed) ; 4(1): 1-15, 2012 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-22202039

RESUMEN

Although the hematopoietic stem cell (HSC) niche has been an active area of study, the concept of the bone marrow microenvironment (BMM) harboring a niche for solid metastatic tumor cells has only recently been considered. The HSC niche and microenvironment that is thought to constitute the solid tumor niche share many of the same structural and functional components, suggesting the possibility that the HSC and tumor niche are one in the same. The osteoblast is a critical component for each of these niches, and is important for regulating cellular processes such homing and migration, growth and survival, and quiescence and dormancy. Current understanding of the HSC niche may provide more insight to better defining the solid tumor niche. As role of the niche in regulating these processes is better understood, new insights to the role of the BMM in metastatic disease may be gained, and provide more potential targets for therapy.


Asunto(s)
Neoplasias/patología , Células Madre Neoplásicas/patología , Nicho de Células Madre , Animales , Humanos , Microambiente Tumoral
16.
Asian J Androl ; 14(3): 423-7, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22367179

RESUMEN

The hematopoietic stem cell (HSC) niche in the bone marrow has been studied extensively over the past few decades, yet the bone marrow microenvironment that supports the growth of metastatic prostate cancer (PCa) has only been recently considered to be a specialized 'niche' as well. New evidence supports the fact that disseminated tumor cells (DTCs) of PCa actually target the HSC niche, displace the occupant HSCs and take up residence in the pre-existing niche space. This review describes some of the evidence and mechanisms by which DTCs act as molecular parasites of the HSC niche. Furthermore, the interactions between DTCs, HSCs and the niche may provide new targets for niche-directed therapy, as well as insight into the perplexing clinical manifestations of metastatic PCa disease.


Asunto(s)
Adenocarcinoma/secundario , Neoplasias de la Médula Ósea/secundario , Células Madre Hematopoyéticas/patología , Neoplasias de la Próstata/patología , Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Neoplasias de la Médula Ósea/tratamiento farmacológico , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Masculino , Metástasis de la Neoplasia , Neoplasia Residual/tratamiento farmacológico , Neoplasia Residual/patología , Osteoblastos/efectos de los fármacos , Osteoblastos/patología , Neoplasias de la Próstata/tratamiento farmacológico
17.
J Clin Invest ; 121(4): 1298-312, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21436587

RESUMEN

HSC homing, quiescence, and self-renewal depend on the bone marrow HSC niche. A large proportion of solid tumor metastases are bone metastases, known to usurp HSC homing pathways to establish footholds in the bone marrow. However, it is not clear whether tumors target the HSC niche during metastasis. Here we have shown in a mouse model of metastasis that human prostate cancer (PCa) cells directly compete with HSCs for occupancy of the mouse HSC niche. Importantly, increasing the niche size promoted metastasis, whereas decreasing the niche size compromised dissemination. Furthermore, disseminated PCa cells could be mobilized out of the niche and back into the circulation using HSC mobilization protocols. Finally, once in the niche, tumor cells reduced HSC numbers by driving their terminal differentiation. These data provide what we believe to be the first evidence that the HSC niche serves as a direct target for PCa during dissemination and plays a central role in bone metastases. Our work may lead to better understanding of the molecular events involved in bone metastases and new therapeutic avenues for an incurable disease.


Asunto(s)
Neoplasias de la Médula Ósea/secundario , Células Madre Hematopoyéticas/patología , Neoplasias de la Próstata , Animales , Neoplasias de la Médula Ósea/patología , Trasplante de Médula Ósea , Línea Celular Tumoral , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Modelos Biológicos , Trasplante de Neoplasias , Osteoblastos/patología , Neoplasias de la Próstata/patología , Donantes de Tejidos , Trasplante Heterólogo
18.
Exp Hematol ; 38(2): 132-40, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19922767

RESUMEN

OBJECTIVE: Despite improvements in current combinational chemotherapy regimens, the prognosis of the (1;19)(q23;p13) translocation (E2A/PBX1)-positive B-cell precursor acute lymphoblastic leukemia (ALL) is poor in pediatric leukemia patients. MATERIALS AND METHODS: In this study, we examined the roles of growth arrest-specific-6 (GAS6)/Mer axis in the interactions between E2A/PBX1-positive B-cell precursor ALL cells and the osteoblastic niche in the bone marrow. RESULTS: Data show that primary human osteoblasts secrete GAS6 in response to the Mer-overexpressed E2A/PBX1-positive ALL cells through mitogen-activated protein kinase signaling pathway and that leukemia cells migrate toward GAS6 using pathways activated by Mer. Importantly, GAS6 supports survival and prevents apoptosis from chemotherapy of E2A/PBX1-positive ALL cells by inducing dormancy. CONCLUSIONS: These data suggest that GAS6/Mer axis regulates homing and survival of the E2A/PBX1-positive B-cell precursor ALL in the bone marrow niche.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/análisis , Médula Ósea/patología , Proteínas de Unión al ADN/análisis , Péptidos y Proteínas de Señalización Intercelular/fisiología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Proteínas Proto-Oncogénicas/análisis , Proteínas Proto-Oncogénicas/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Quimiotaxis , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/farmacología , Masculino , Osteoblastos/metabolismo , Factor de Transcripción 1 de la Leucemia de Células Pre-B , Leucemia-Linfoma Linfoblástico de Células Precursoras B/enzimología , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Tirosina Quinasa c-Mer
19.
Neoplasia ; 12(2): 116-27, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20126470

RESUMEN

Our recent studies have shown that annexin II, expressed on the cell surface of osteoblasts, plays an important role in the adhesion of hematopoietic stem cells (HSCs) to the endosteal niche. Similarly, prostate cancer (PCa) cells express the annexin II receptor and seem to use the stem cell niche for homing to the bone marrow. The role of the niche is thought to be the induction and sustenance of HSC dormancy. If metastatic PCa cells occupy a similar or the same ecological niche as HSCs, then it is likely that the initial role of the HSC niche will be to induce dormancy in metastatic cells. In this study, we demonstrate that the binding of PCa to annexin II induces the expression of the growth arrest-specific 6 (GAS6) receptors AXL, Sky, and Mer, which, in the hematopoietic system, induce dormancy. In addition, GAS6 produced by osteoblasts prevents PCa proliferation and protects PCa from chemotherapy-induced apoptosis. Our results suggest that the activation of GAS6 receptors on PCa in the bone marrow environment may play a critical role as a molecular switch, establishing metastatic tumor cell dormancy.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Invasividad Neoplásica , Neoplasias de la Próstata/metabolismo , Nicho de Células Madre/metabolismo , Animales , Anexina A2/metabolismo , Apoptosis/fisiología , Western Blotting , Línea Celular Tumoral , Proliferación Celular , Separación Celular , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Inmunohistoquímica , Masculino , Ratones , Osteoblastos/metabolismo , Neoplasias de la Próstata/patología , Proteínas Tirosina Quinasas Receptoras , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Nicho de Células Madre/citología , Análisis de Matrices Tisulares
20.
PLoS One ; 5(5): e10853, 2010 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-20523730

RESUMEN

BACKGROUND: It is well established that bleeding activates the hematopoietic system to regenerate the loss of mature blood elements. We have shown that hematopoietic stem cells (HSCs) isolated from animals challenged with an acute bleed regulate osteoblast differentiation from marrow stromal cells. This suggests that HSCs participate in bone formation where the molecular basis for this activity is the production of BMP2 and BMP6 by HSCs. Yet, what stimulates HSCs to produce BMPs is unclear. METHODOLOGY/PRINCIPAL FINDINGS: In this study, we demonstrate that erythropoietin (Epo) activates Jak-Stat signaling pathways in HSCs which leads to the production of BMPs. Critically, Epo also directly activates mesenchymal cells to form osteoblasts in vitro, which in vivo leads to bone formation. Importantly, Epo first activates osteoclastogenesis which is later followed by osteoblastogenesis that is induced by either Epo directly or the expression of BMPs by HSCs to form bone. CONCLUSIONS/SIGNIFICANCE: These data for the first time demonstrate that Epo regulates the formation of bone by both direct and indirect pathways, and further demonstrates the exquisite coupling between hematopoiesis and osteopoiesis in the marrow.


Asunto(s)
Eritropoyetina/metabolismo , Hematopoyesis , Osteogénesis , Animales , Animales Recién Nacidos , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Eritropoyetina/farmacología , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteogénesis/efectos de los fármacos , Fenotipo , Receptores de Eritropoyetina/metabolismo , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA