Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cell Rep ; 12(6): 992-1005, 2015 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-26235614

RESUMO

Several questions about the role of the oxygen sensor prolyl-hydroxylase 2 (PHD2) in cancer have not been addressed. First, the role of PHD2 in metastasis has not been studied in a spontaneous tumor model. Here, we show that global PHD2 haplodeficiency reduced metastasis without affecting tumor growth. Second, it is unknown whether PHD2 regulates cancer by affecting cancer-associated fibroblasts (CAFs). We show that PHD2 haplodeficiency reduced metastasis via two mechanisms: (1) by decreasing CAF activation, matrix production, and contraction by CAFs, an effect that surprisingly relied on PHD2 deletion in cancer cells, but not in CAFs; and (2) by improving tumor vessel normalization. Third, the effect of concomitant PHD2 inhibition in malignant and stromal cells (mimicking PHD2 inhibitor treatment) is unknown. We show that global PHD2 haplodeficiency, induced not only before but also after tumor onset, impaired metastasis. These findings warrant investigation of PHD2's therapeutic potential.


Assuntos
Fibroblastos/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Neoplasias/metabolismo , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Prolina Dioxigenases do Fator Induzível por Hipóxia/genética , Immunoblotting , Imuno-Histoquímica , Masculino , Camundongos , Modelos Biológicos , Metástase Neoplásica , Neoplasias/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
2.
Neoplasia ; 14(10): 943-51, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23097628

RESUMO

Carcinoma-associated fibroblasts are key contributors of the tumor microenvironment that regulates carcinoma progression. They consist of a heterogeneous cell population with diverse origins, phenotypes, and functions. In the present report, we have explored the contribution of bone marrow (BM)-derived cells to generate different fibroblast subsets that putatively produce the matrix metalloproteinase 13 (MMP13) and affect cancer cell invasion. A murine model of skin carcinoma was applied to mice, irradiated, and engrafted with BM isolated from green fluorescent protein (GFP) transgenic mice. We provide evidence that one third of BM-derived GFP(+) cells infiltrating the tumor expressed the chondroitin sulfate proteoglycan NG2 (pericytic marker) or α-smooth muscle actin (α-SMA, myofibroblast marker), whereas almost 90% of Thy1(+) fibroblasts were originating from resident GFP-negative cells. MMP13producing cells were exclusively α-SMA(+) cells and derived from GFP(+) BM cells. To investigate their impact on tumor invasion, we isolated mesenchymal stem cells (MSCs) from the BM of wild-type and MMP13-deficient mice. Wild-type MSC promoted cancer cell invasion in a spheroid assay, whereas MSCs obtained from MMP13-deficient mice failed to. Our data support the concept of fibroblast subset specialization with BM-derived α-SMA(+) cells being the main source of MMP13, a stromal mediator of cancer cell invasion.


Assuntos
Medula Óssea/patologia , Fibroblastos/patologia , Metaloproteinase 13 da Matriz/fisiologia , Células-Tronco Mesenquimais/patologia , Miofibroblastos/patologia , Neoplasias/patologia , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Medula Óssea/metabolismo , Feminino , Fibroblastos/metabolismo , Citometria de Fluxo , Humanos , Técnicas Imunoenzimáticas , Hibridização In Situ , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miofibroblastos/metabolismo , Invasividade Neoplásica , Neoplasias/genética , Neoplasias/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas , Microambiente Tumoral
3.
Blood ; 119(21): 5048-56, 2012 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-22490679

RESUMO

Lymphatic dysfunctions are associated with several human diseases, including lymphedema and metastatic spread of cancer. Although it is well recognized that lymphatic capillaries attach directly to interstitial matrix mainly composed of fibrillar type I collagen, the interactions occurring between lymphatics and their surrounding matrix have been overlooked. In this study, we demonstrate how matrix metalloproteinase (MMP)-2 drives lymphatic morphogenesis through Mmp2-gene ablation in mice, mmp2 knockdown in zebrafish and in 3D-culture systems, and through MMP2 inhibition. In all models used in vivo (3 murine models and thoracic duct development in zebrafish) and in vitro (lymphatic ring and spheroid assays), MMP2 blockage or down-regulation leads to reduced lymphangiogenesis or altered vessel branching. Our data show that lymphatic endothelial cell (LEC) migration through collagen fibers is affected by physical matrix constraints (matrix composition, density, and cross-linking). Transmission electron microscopy and confocal reflection microscopy using DQ-collagen highlight the contribution of MMP2 to mesenchymal-like migration of LECs associated with collagen fiber remodeling. Our findings provide new mechanistic insight into how LECs negotiate an interstitial type I collagen barrier and reveal an unexpected MMP2-driven collagenolytic pathway for lymphatic vessel formation and morphogenesis.


Assuntos
Linfangiogênese/genética , Vasos Linfáticos/embriologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 2 da Matriz/fisiologia , Animais , Animais Geneticamente Modificados , Células Cultivadas , Colágeno Tipo I/metabolismo , Colagenases/genética , Colagenases/metabolismo , Colagenases/fisiologia , Embrião não Mamífero , Líquido Extracelular/enzimologia , Líquido Extracelular/metabolismo , Feminino , Humanos , Vasos Linfáticos/metabolismo , Vasos Linfáticos/fisiologia , Masculino , Metaloproteinase 2 da Matriz/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peixe-Zebra
4.
Nature ; 479(7371): 122-6, 2011 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-21983962

RESUMO

PHD2 serves as an oxygen sensor that rescues blood supply by regulating vessel formation and shape in case of oxygen shortage. However, it is unknown whether PHD2 can influence arteriogenesis. Here we studied the role of PHD2 in collateral artery growth by using hindlimb ischaemia as a model, a process that compensates for the lack of blood flow in case of major arterial occlusion. We show that Phd2 (also known as Egln1) haplodeficient (Phd2(+/-)) mice displayed preformed collateral arteries that preserved limb perfusion and prevented tissue necrosis in ischaemia. Improved arteriogenesis in Phd2(+/-) mice was due to an expansion of tissue-resident, M2-like macrophages and their increased release of arteriogenic factors, leading to enhanced smooth muscle cell (SMC) recruitment and growth. Both chronic and acute deletion of one Phd2 allele in macrophages was sufficient to skew their polarization towards a pro-arteriogenic phenotype. Mechanistically, collateral vessel preconditioning relied on the activation of canonical NF-κB pathway in Phd2(+/-) macrophages. These results unravel how PHD2 regulates arteriogenesis and artery homeostasis by controlling a specific differentiation state in macrophages and suggest new treatment options for ischaemic disorders.


Assuntos
Artérias/crescimento & desenvolvimento , Extremidades/irrigação sanguínea , Isquemia/prevenção & controle , Macrófagos/metabolismo , Pró-Colágeno-Prolina Dioxigenase/deficiência , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Alelos , Animais , Modelos Animais de Doenças , Extremidades/patologia , Feminino , Heterozigoto , Homeostase , Prolina Dioxigenases do Fator Induzível por Hipóxia , Isquemia/patologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Miócitos de Músculo Liso/citologia , NF-kappa B/metabolismo , Necrose , Fenótipo , Pró-Colágeno-Prolina Dioxigenase/genética
5.
BMC Cell Biol ; 12: 29, 2011 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-21702933

RESUMO

BACKGROUND: Abnormal lymphatic vessel formation (lymphangiogenesis) is associated with different pathologies such as cancer, lymphedema, psoriasis and graft rejection. Lymphatic vasculature displays distinctive features than blood vasculature, and mechanisms underlying the formation of new lymphatic vessels during physiological and pathological processes are still poorly documented. Most studies on lymphatic vessel formation are focused on organism development rather than lymphangiogenic events occurring in adults. We have here studied lymphatic vessel formation in two in vivo models of pathological lymphangiogenesis (corneal assay and lymphangioma). These data have been confronted to those generated in the recently set up in vitro model of lymphatic ring assay. Ultrastructural analyses through Transmission Electron Microscopy (TEM) were performed to investigate tube morphogenesis, an important differentiating process observed during endothelial cell organization into capillary structures. RESULTS: In both in vivo models (lymphangiogenic corneal assay and lymphangioma), migrating lymphatic endothelial cells extended long processes exploring the neighboring environment and organized into cord-like structures. Signs of intense extracellular matrix remodeling were observed extracellularly and inside cytoplasmic vacuoles. The formation of intercellular spaces between endothelial cells led to tube formation. Proliferating lymphatic endothelial cells were detected both at the tips of sprouting capillaries and inside extending sprouts. The different steps of lymphangiogenesis observed in vivo are fully recapitulated in vitro, in the lymphatic ring assay and include: (1) endothelial cell alignment in cord like structure, (2) intracellular vacuole formation and (3) matrix degradation. CONCLUSIONS: In this study, we are providing evidence for lymphatic vessel formation through tunneling relying on extensive matrix remodeling, migration and alignment of sprouting endothelial cells into tubular structures. In addition, our data emphasize the suitability of the lymphatic ring assay to unravel mechanisms underlying lymphangiogenesis.


Assuntos
Linfangiogênese/fisiologia , Vasos Linfáticos/fisiologia , Animais , Movimento Celular , Células Cultivadas , Córnea/patologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Endotélio Linfático/metabolismo , Endotélio Linfático/patologia , Feminino , Adjuvante de Freund , Hiperplasia , Técnicas In Vitro , Linfangioma/etiologia , Linfangioma/patologia , Vasos Linfáticos/patologia , Vasos Linfáticos/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão
6.
Int J Cancer ; 128(2): 283-93, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20232379

RESUMO

Angiogenesis, extracellular matrix remodeling and cell migration are associated with cancer progression and involve at least, the plasminogen activating system and its main physiological inhibitor, the plasminogen activator inhibitor-1 (PAI-1). Considering the recognized importance of PAI-1 in the regulation of tumor angiogenesis and invasion in murine models of skin tumor transplantation, we explored the functional significance of PAI-1 during early stages of neoplastic progression in the transgenic mouse model of multistage epithelial carcinogenesis (K14-HPV16 mice). We have studied the effect of genetic deletion of PAI-1 on inflammation, angiogenesis, lymphangiogenesis and tumor progression. In this model, PAI-1 deficiency neither impaired keratinocyte hyperproliferation or tumor development nor affected the infiltration of inflammatory cells and development of angiogenic or lymphangiogenic vasculature. We are reporting evidence for concomitant lymphangiogenic and angiogenic switches independent to PAI-1 status. Taken together, these data indicate that PAI-1 is not rate limiting for neoplastic progression and vascularization during premalignant progression, or that there is a functional redundancy between PAI-1 and other tumor regulators, masking the effect of PAI-1 deficiency in this long-term model of multistage epithelial carcinogenesis.


Assuntos
Papillomavirus Humano 16/genética , Queratina-14/genética , Inibidor 1 de Ativador de Plasminogênio/fisiologia , Neoplasias Cutâneas/etiologia , Animais , Linfangiogênese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Inibidor 1 de Ativador de Plasminogênio/deficiência , Lesões Pré-Cancerosas/etiologia , Neoplasias Cutâneas/irrigação sanguínea
7.
PLoS One ; 5(3): e9653, 2010 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-20300183

RESUMO

The purpose of this study is to explore the function of plasminogen activator inhibitor-1 (PAI-1) during pathological lymphangiogenesis. PAI-1, the main physiological inhibitor of plasminogen activators is involved in pathological angiogenesis at least by controlling extracellular proteolysis and by regulating endothelial cell survival and migration. Protease system's role in lymphangiogenesis is unknown yet. Thus, based on its important pro-angiogenic effect, we hypothesized that PAI-1 may regulate lymphangiogenesis associated at least with metastatic dissemination of cancer cells. To address this issue, we studied the impact of PAI-1 deficiency in various murine models of tumoral lymphangiogenesis. Wild-type PAI-1 proficient mice were used as controls. We provide for the first time evidence that PAI-1 is dispensable for tumoral lymphangiogenesis associated with breast cancers either induced by mammary carcinoma cell injection or spontaneously appearing in transgenic mice expressing the polyomavirus middle T antigen (PymT) under the control of a mouse mammary tumor virus long-terminal repeat promoter (MMTV-LTR). We also investigated inflammation-related lymphatic vessel recruitment by using two inflammatory models. PAI-1 deficiency did neither affect the development of lymphangioma nor burn-induced corneal lymphangiogenesis. These novel data suggest that vascular remodelling associated with lymphangiogenesis and angiogenesis involve different molecular determinants. PAI-1 does not appear as a potential therapeutic target to counteract pathological lymphangiogenesis.


Assuntos
Inibidor 1 de Ativador de Plasminogênio/fisiologia , Animais , Antígenos Transformantes de Poliomavirus/metabolismo , Linhagem Celular Tumoral , Feminino , Heterozigoto , Humanos , Inflamação , Linfangioma/metabolismo , Masculino , Vírus do Tumor Mamário do Camundongo/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Metástase Neoplásica
8.
FASEB J ; 24(1): 8-21, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19726757

RESUMO

Lymphangiogenesis, the formation of new lymphatic vessels from preexisting ones, is an important biological process associated with diverse pathologies, such as metastatic dissemination and graft rejection. In addition, lymphatic hypoplasia characterizes lymphedema, usually a progressive and lifelong condition for which no curative treatment exists. Much progress has been made in recent years in identifying molecules specifically expressed on lymphatic vessels and in the setting up of in vitro and in vivo models of lymphangiogenesis. These new tools rapidly provided an abundance of information on the mechanisms underlying lymphatic development and the progression of diseases associated with lymphatic dysfunction. In this review, we describe the common in vitro and in vivo models of lymphangiogenesis that have proven suitable for investigating lymphatic biology and the interactions occurring between lymphatic vessels and other cells, such as immune cells and cancer cells. Their rationales and limitations are discussed and illustrated by the most informative findings obtained with them.


Assuntos
Linfangiogênese/fisiologia , Modelos Biológicos , Animais , Técnicas de Cultura de Células , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/citologia , Humanos , Linfangioma/etiologia , Metástase Linfática/fisiopatologia , Vasos Linfáticos/citologia , Vasos Linfáticos/embriologia , Vasos Linfáticos/fisiologia , Linfedema/etiologia , Técnicas de Cultura de Órgãos
9.
J Pathol ; 219(3): 356-64, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19718705

RESUMO

Vascular endothelial growth factor-D (VEGF-D) is one of the two ligands of the VEGFR-3 receptor on lymphatic endothelial cells. Gene-silencing studies in mice and Xenopus tadpoles recently showed that the role of endogenous VEGF-D in lymphatic development is moderate. By contrast, exogenous VEGF-D is capable of stimulating lymphangiogenesis. Nonetheless, its endogenous role in pathological conditions remains largely unknown. Hence, we reassessed its role in disease, using Vegf-d(null) mice. Vegf-d(null) mice were generated that, under physiological conditions, displayed normal embryonic and postnatal lymphangiogenesis and lymphatic remodelling, efficient lymphatic functioning and normal health. Vegf-d(null) mice also reponded normally in models of skin wound healing and healing of infarcted myocardium, despite enhanced expression of VEGF-D in these models in wild-type mice. In contrast, Vegf-d(null) mice displayed reduced peritumoral lymphangiogenesis and lymph node metastasis in an orthotopic pancreatic tumour model. Together, our data indicate that endogenous VEGF-D in mice is dispensible for lymphangiogenesis during development, in postnatal and adult physiology and in several pathological conditions, but significantly contributes to lymphatic metastasis.


Assuntos
Linfangiogênese/fisiologia , Metástase Linfática/fisiopatologia , Fator D de Crescimento do Endotélio Vascular/deficiência , Adenocarcinoma/metabolismo , Adenocarcinoma/secundário , Animais , Modelos Animais de Doenças , Desenvolvimento Embrionário/fisiologia , Feminino , Marcação de Genes/métodos , Neoplasias Hepáticas/metabolismo , Linfangioma/metabolismo , Sistema Linfático/embriologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infarto do Miocárdio/fisiopatologia , Transplante de Neoplasias , Neoplasias Pancreáticas/metabolismo , Fenótipo , Pele/lesões , Fator D de Crescimento do Endotélio Vascular/fisiologia , Cicatrização/fisiologia
10.
J Cell Mol Med ; 13(9B): 4002-13, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19426156

RESUMO

The present study aims at investigating the mechanism by which membrane-type 4 matrix metalloproteinase (MT4-MMP), a membrane-anchored MMP expressed by human breast tumour cells promotes the metastatic dissemination into lung. We applied experimental (intravenous) and spontaneous (subcutaneous) models of lung metastasis using human breast adenocarcinoma MDA-MB-231 cells overexpressing or not MT4-MMP. We found that MT4-MMP does not affect lymph node colonization nor extravasation of cells from the bloodstream, but increases the intravasation step leading to metastasis. Ultrastructural and fluorescent microscopic observations coupled with automatic computer-assisted quantifications revealed that MT4-MMP expression induces blood vessel enlargement and promotes the detachment of mural cells from the vascular tree, thus causing an increased tumour vascular leak. On this basis, we propose that MT4-MMP promotes lung metastasis by disturbing the tumour vessel integrity and thereby facilitating tumour cell intravasation.


Assuntos
Neoplasias da Mama/enzimologia , Neoplasias Pulmonares/enzimologia , Metaloproteinase 17 da Matriz/metabolismo , Animais , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Humanos , Neoplasias Pulmonares/secundário , Metástase Linfática , Neoplasias Mamárias Animais/irrigação sanguínea , Neoplasias Mamárias Animais/enzimologia , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Nus , Microscopia Eletrônica de Transmissão/métodos , Microscopia de Fluorescência/métodos , Transplante de Neoplasias , Neovascularização Patológica , Pericitos/metabolismo
11.
Int J Cancer ; 125(1): 34-42, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19326439

RESUMO

The phloroglucinol derivative hyperforin, a major bioactive constituent of St. John's wort, is increasingly recognized as being able to regulate a variety of pathobiological processes and, thus, to possess potential therapeutic properties. In the context of cancer, hyperforin induces the apoptosis of cancer cells, inhibits angiogenesis and suppresses metastasis formation. Here, we report a new pharmacological function of hyperforin and its stabilized derivative aristoforin, namely the suppression of lymphatic endothelial cell (LEC) growth and lymphangiogenesis. At concentrations less than 10 microM, we found that these compounds induce cell cycle arrest of LECs, and at higher concentrations induce apoptosis. The loss of mitochondrial membrane potential and the activation of caspase-9 during the induction of apoptosis indicate that the intrinsic pathway of apoptosis is stimulated by these compounds, similar to the situation in tumor cells. In thoracic duct ring outgrowth assays, hyperforin and aristoforin both inhibited lymphangiogenesis, as evidenced by the suppression of lymphatic capillary outgrowth. In an in vivo animal model, both compounds were able to inhibit tumor-induced lymphangiogenesis. Together these data substantiate a new role for hyperforin and its derivatives as suppressors of lymphangiogenesis, and support their further investigation as potential anticancer drugs that target tumor growth and metastasis at multiple levels.


Assuntos
Proliferação de Células/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Linfangiogênese/efeitos dos fármacos , Neoplasias Experimentais/patologia , Floroglucinol/análogos & derivados , Terpenos/farmacologia , Animais , Aorta Torácica/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Compostos Bicíclicos com Pontes/farmacologia , Caspase 3/metabolismo , Caspase 8/metabolismo , Ciclo Celular/efeitos dos fármacos , Células Cultivadas , Citocromos c/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Feminino , Humanos , Técnicas In Vitro , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Floroglucinol/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Wistar , Ensaios Antitumorais Modelo de Xenoenxerto
12.
BMC Neurosci ; 5: 33, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15369599

RESUMO

BACKGROUND: Spontaneous repair is limited after CNS injury or degeneration because neurogenesis and axonal regrowth rarely occur in the adult brain. As a result, cell transplantation has raised much interest as potential treatment for patients with CNS lesions. Several types of cells have been considered as candidates for such cell transplantation and replacement therapies. Foetal brain tissue has already been shown to have significant effects in patients with Parkinson's disease. Clinical use of the foetal brain tissue is, however, limited by ethical and technical problems as it requires high numbers of grafted foetal cells and immunosuppression. Alternatively, several reports suggested that mesenchymal stem cells, isolated from adult bone marrow, are multipotent cells and could be used in autograft approach for replacement therapies. RESULTS: In this study, we addressed the question of the possible influence of mesenchymal stem cells on neural stem cell fate. We have previously reported that adult rat mesenchymal stem cells are able to express nestin in defined culture conditions (in the absence of serum and after 25 cell population doublings) and we report here that nestin-positive (but not nestin-negative) mesenchymal stem cells are able to favour the astroglial lineage in neural progenitors and stem cells cultivated from embryonic striatum. The increase of the number of GFAP-positive cells is associated with a significant decrease of the number of Tuj1- and O4-positive cells. Using quantitative RT-PCR, we demonstrate that mesenchymal stem cells express LIF, CNTF, BMP2 and BMP4 mRNAs, four cytokines known to play a role in astroglial fate decision. In this model, BMP4 is responsible for the astroglial stimulation and oligodendroglial inhibition, as 1) this cytokine is present in a biologically-active form only in nestin-positive mesenchymal stem cells conditioned medium and 2) anti-BMP4 antibodies inhibit the nestin-positive mesenchymal stem cells conditioned medium inducing effect on astrogliogenesis. CONCLUSIONS: When thinking carefully about mesenchymal stem cells as candidates for cellular therapy in neurological diseases, their effects on resident neural cell fate have to be considered.


Assuntos
Astrócitos/citologia , Proteínas Morfogenéticas Ósseas/fisiologia , Proteínas de Filamentos Intermediários/biossíntese , Células-Tronco Mesenquimais/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Neurônios/citologia , Células-Tronco/metabolismo , Animais , Antígenos de Diferenciação/biossíntese , Células da Medula Óssea/citologia , Proteína Morfogenética Óssea 4 , Proteínas Morfogenéticas Ósseas/biossíntese , Proteínas Morfogenéticas Ósseas/metabolismo , Contagem de Células , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Divisão Celular , Linhagem da Célula , Células Cultivadas , Técnicas de Cocultura , Meios de Cultivo Condicionados/farmacologia , Citocinas/genética , Proteína Glial Fibrilar Ácida/biossíntese , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos , Nestina , RNA Mensageiro/biossíntese , Ratos , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA