Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Respir Res ; 22(1): 265, 2021 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-34666752

RESUMO

RATIONALE: αv integrins, key regulators of transforming growth factor-ß activation and fibrogenesis in in vivo models of pulmonary fibrosis, are expressed on abnormal epithelial cells (αvß6) and fibroblasts (αvß1) in fibrotic lungs. OBJECTIVES: We evaluated multiple αv integrin inhibition strategies to assess which most effectively reduced fibrogenesis in explanted lung tissue from patients with idiopathic pulmonary fibrosis. METHODS: Selective αvß6 and αvß1, dual αvß6/αvß1, and multi-αv integrin inhibitors were characterized for potency, selectivity, and functional activity by ligand binding, cell adhesion, and transforming growth factor-ß cell activation assays. Precision-cut lung slices generated from lung explants from patients with idiopathic pulmonary fibrosis or bleomycin-challenged mouse lungs were treated with integrin inhibitors or standard-of-care drugs (nintedanib or pirfenidone) and analyzed for changes in fibrotic gene expression or TGF-ß signaling. Bleomycin-challenged mice treated with dual αvß6/αvß1 integrin inhibitor, PLN-74809, were assessed for changes in pulmonary collagen deposition and Smad3 phosphorylation. MEASUREMENTS AND MAIN RESULTS: Inhibition of integrins αvß6 and αvß1 was additive in reducing type I collagen gene expression in explanted lung tissue slices from patients with idiopathic pulmonary fibrosis. These data were replicated in fibrotic mouse lung tissue, with no added benefit observed from inhibition of additional αv integrins. Antifibrotic efficacy of dual αvß6/αvß1 integrin inhibitor PLN-74809 was confirmed in vivo, where dose-dependent inhibition of pulmonary Smad3 phosphorylation and collagen deposition was observed. PLN-74809 also, more potently, reduced collagen gene expression in fibrotic human and mouse lung slices than clinically relevant concentrations of nintedanib or pirfenidone. CONCLUSIONS: In the fibrotic lung, dual inhibition of integrins αvß6 and αvß1 offers the optimal approach for blocking fibrogenesis resulting from integrin-mediated activation of transforming growth factor-ß.


Assuntos
Antifibróticos/farmacologia , Células Epiteliais/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fibrose Pulmonar Idiopática/tratamento farmacológico , Integrina alfa6beta1/antagonistas & inibidores , Pulmão/efeitos dos fármacos , Receptores de Vitronectina/antagonistas & inibidores , Animais , Bleomicina , Linhagem Celular , Técnicas de Cocultura , Cadeia alfa 1 do Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I/metabolismo , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Fibrose Pulmonar Idiopática/genética , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Integrina alfa6beta1/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Camundongos Endogâmicos C57BL , Fosforilação , Receptores de Vitronectina/metabolismo , Transdução de Sinais , Proteína Smad3/metabolismo
2.
Hepatology ; 65(1): 78-88, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27706836

RESUMO

Excess collagen synthesis (fibrogenesis) in the liver plays a causal role in the progression of nonalcoholic fatty liver disease (NAFLD). Methods are needed to identify patients with more rapidly progressing disease and to demonstrate early response to treatment. We describe here a novel method to quantify hepatic fibrogenesis flux rates both directly in liver tissue and noninvasively in blood. Twenty-one patients with suspected NAFLD ingested heavy water (2 H2 O, 50-mL aliquots) two to three times daily for 3-5 weeks prior to a clinically indicated liver biopsy. Liver collagen fractional synthesis rate (FSR) and plasma lumican FSR were measured based on 2 H labeling using tandem mass spectrometry. Patients were classified by histology for fibrosis stage (F0-F4) and as having nonalcoholic fatty liver or nonalcoholic steatohepatitis (NASH). Magnetic resonance elastography measurements of liver stiffness were also performed. Hepatic collagen FSR in NAFLD increased with advancing disease stage (e.g., higher in NASH than nonalcoholic fatty liver, positive correlation with fibrosis score and liver stiffness) and correlated with hemoglobin A1C. In addition, plasma lumican FSR demonstrated a significant correlation with hepatic collagen FSR. CONCLUSION: Using a well-characterized cohort of patients with biopsy-proven NAFLD, this study demonstrates that hepatic scar in NASH is actively remodeled even in advanced fibrosis, a disease that is generally regarded as static and slowly progressive. Moreover, hepatic collagen FSR correlates with established risks for fibrotic disease progression in NASH, and plasma lumican FSR correlates with hepatic collagen FSR, suggesting applications as direct or surrogate markers, respectively, of hepatic fibrogenesis in humans. (Hepatology 2017;65:78-88).


Assuntos
Cirrose Hepática/sangue , Cirrose Hepática/patologia , Biópsia , Colágeno/metabolismo , Progressão da Doença , Matriz Extracelular/metabolismo , Feminino , Humanos , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática/complicações , Lumicana/sangue , Masculino , Pessoa de Meia-Idade , Hepatopatia Gordurosa não Alcoólica/complicações
3.
PLoS One ; 10(4): e0123311, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25909381

RESUMO

Accumulation and degradation of scar tissue in fibrotic liver disease occur slowly, typically over many years. Direct measurement of fibrogenesis, the rate of scar tissue deposition, may provide valuable therapeutic and prognostic information. We describe here results from a pilot study utilizing in vivo metabolic labeling to measure the turnover rate of hepatic collagen and collagen-associated proteins in plasma for the first time in human subjects. Eight subjects with chronic liver disease were labeled with daily oral doses of 2H2O for up to 8 weeks prior to diagnostic liver biopsy and plasma collection. Tandem mass spectrometry was used to measure the abundance and fractional synthesis rate (FSR) of proteins in liver and blood. Relative protein abundance and FSR data in liver revealed marked differences among subjects. FSRs of hepatic type I and III collagen ranged from 0.2-0.6% per day (half-lives of 4 months to a year) and correlated significantly with worsening histologic fibrosis. Analysis of plasma protein turnover revealed two collagen-associated proteins, lumican and transforming growth factor beta-induced-protein (TGFBI), exhibiting FSRs that correlated significantly with FSRs of hepatic collagen. In summary, this is the first direct measurement of liver collagen turnover in vivo in humans and suggests a high rate of collagen remodeling in advanced fibrosis. In addition, the FSRs of collagen-associated proteins in plasma are measurable and may provide a novel strategy for monitoring hepatic fibrogenesis rates.


Assuntos
Proteínas de Transporte/sangue , Colágeno/metabolismo , Hepatopatias/metabolismo , Hepatopatias/patologia , Adulto , Idoso , Biópsia , Análise por Conglomerados , Feminino , Humanos , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática/sangue , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Hepatopatias/sangue , Hepatopatias/diagnóstico , Hepatopatias/etiologia , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Ligação Proteica , Proteoma , Proteômica/métodos , Fator de Crescimento Transformador beta1/metabolismo
4.
Mol Cell Proteomics ; 13(7): 1741-52, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24741116

RESUMO

Fibrotic disease is characterized by the pathological accumulation of extracellular matrix (ECM) proteins. Surprisingly, very little is known about the synthesis and degradation rates of the many proteins and proteoglycans that constitute healthy or pathological extracellular matrix. A comprehensive understanding of altered ECM protein synthesis and degradation during the onset and progression of fibrotic disease would be immensely valuable. We have developed a dynamic proteomics platform that quantifies the fractional synthesis rates of large numbers of proteins via stable isotope labeling and LC/MS-based mass isotopomer analysis. Here, we present the first broad analysis of ECM protein kinetics during the onset of experimental pulmonary fibrosis. Mice were labeled with heavy water for up to 21 days following the induction of lung fibrosis with bleomycin. Lung tissue was subjected to sequential protein extraction to fractionate cellular, guanidine-soluble ECM proteins and residual insoluble ECM proteins. Fractional synthesis rates were calculated for 34 ECM proteins or protein subunits, including collagens, proteoglycans, and microfibrillar proteins. Overall, fractional synthesis rates of guanidine-soluble ECM proteins were faster than those of insoluble ECM proteins, suggesting that the insoluble fraction reflected older, more mature matrix components. This was confirmed through the quantitation of pyridinoline cross-links in each protein fraction. In fibrotic lung tissue, there was a significant increase in the fractional synthesis of unique sets of matrix proteins during early (pre-1 week) and late (post-1 week) fibrotic response. Furthermore, we isolated fast turnover subpopulations of several ECM proteins (e.g. type I collagen) based on guanidine solubility, allowing for accelerated detection of increased synthesis of typically slow-turnover protein populations. This establishes the presence of multiple kinetic pools of pulmonary collagen in vivo with altered turnover rates during evolving fibrosis. These data demonstrate the utility of dynamic proteomics in analyzing changes in ECM protein turnover associated with the onset and progression of fibrotic disease.


Assuntos
Colágeno Tipo I/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Fibrose Pulmonar/patologia , Animais , Membrana Basal/metabolismo , Bleomicina/farmacologia , Colágeno Tipo I/biossíntese , Óxido de Deutério , Matriz Extracelular/metabolismo , Cromatografia Gasosa-Espectrometria de Massas , Perfilação da Expressão Gênica , Marcação por Isótopo , Camundongos , Camundongos Endogâmicos C57BL , Microfibrilas/metabolismo , Proteoglicanas/metabolismo , Proteômica , Fibrose Pulmonar/induzido quimicamente
5.
FASEB J ; 27(12): 4844-52, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24005905

RESUMO

Alginate hydrogels containing cell-instructive cues are the subject of intense interest for their use as cell carriers in bone tissue engineering. Peptides and proteins are chemically grafted onto these hydrophilic materials to facilitate adhesion and direct phenotype of entrapped cells. However, the presentation of a single or small number of peptides does not represent the complexity of the native extracellular matrix (ECM) of bony tissues. Mesenchymal stem cells (MSCs) secrete ECM that can be harvested and deposited on various substrata to promote osteogenic differentiation. In this study, we hypothesized that the presentation of engineered cell-secreted ECM on microbeads suspended in alginate hydrogels would promote cell adhesion and enhance osteogenic differentiation of undifferentiated MSCs without chemical incorporation of cell-adhesive peptides. Human MSCs entrapped in alginate hydrogels loaded with ECM-coated beads showed increased interaction with beads, when compared with cells suspended in hydrogels containing uncoated blank (BLK) beads. MSCs entrapped in ECM gels exhibited increased alkaline phosphatase (ALP) activity and expression of osteogenic genes in vitro compared with hydrogels modified with arginine-glycine-aspartic acid (RGD)-containing peptides. Transplantation of MSCs into an ectopic site resulted in significant increases in blood vessel density for ECM hydrogels when compared with the BLK or RGD gels. Furthermore, we observed comparable levels of bone formation at 6 wk with ECM and RGD hydrogels. These findings demonstrate that engineered ECM can be deployed in a minimally invasive manner to direct the formation of bony tissue. This strategy may provide an alternative to the engraftment of proteins or peptides onto the polymer backbone of hydrogels for directing cellular behavior.


Assuntos
Alginatos/farmacologia , Materiais Biocompatíveis/farmacologia , Hidrogéis/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Microesferas , Osteogênese/efeitos dos fármacos , Fosfatase Alcalina/metabolismo , Animais , Materiais Biocompatíveis/química , Matriz Extracelular/metabolismo , Humanos , Hidrogéis/química , Masculino , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Oligopeptídeos/farmacologia , Ratos
6.
Tissue Eng Part A ; 18(19-20): 2148-57, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22651377

RESUMO

Cells in culture deposit a complex extracellular matrix that remains intact following decellularization and possesses the capacity to modulate cell phenotype. The direct application of such decellularized matrices (DMs) to 3D substrates is problematic, as transport issues influence the homogeneous deposition, decellularization, and modification of DM surface coatings. In an attempt to address this shortcoming, we hypothesized that DMs deposited by human mesenchymal stem cells (MSCs) could be transferred to the surface of polymeric scaffolds while maintaining their capacity to direct cell fate. The ability of the transferred DM (tDM)-coated scaffolds to enhance the osteogenic differentiation of undifferentiated and osteogenically induced MSCs under osteogenic conditions in vitro was confirmed. tDM-coated scaffolds increased MSC expression of osteogenic marker genes (BGLAP, IBSP) and intracellular alkaline phosphatase production. In addition, undifferentiated MSCs deposited significantly more calcium when seeded onto tDM-coated scaffolds compared with control scaffolds. MSC-seeded tDM-coated scaffolds subcutaneously implanted in nude rats displayed significantly higher blood vessel density after 2 weeks compared with cells on uncoated scaffolds, but we did not observe significant differences in mineral deposition after 8 weeks. These data demonstrate that DM-coatings produced in 2D culture can be successfully transferred to 3D substrates and retain their capacity to modulate cell phenotype.


Assuntos
Células-Tronco Mesenquimais/citologia , Células Cultivadas , Humanos , Osteogênese/fisiologia , Polímeros , Alicerces Teciduais
7.
Tissue Eng Part A ; 18(13-14): 1520-8, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22546052

RESUMO

Mesenchymal stem cells (MSCs) seeded in composite implants formed of hydroxyapatite (HA) and poly (lactide-co-glycolide) (PLG) exhibit increased osteogenesis and enhanced angiogenic potential. Endothelial colony-forming cells (ECFCs) can participate in de novo vessel formation when implanted in vivo. The aim of this study was to determine the capacity of HA-PLG composites to cotransplant MSCs and ECFCs, with the goal of accelerating vascularization and resultant bone formation. The incorporation of HA into PLG scaffolds improved the efficiency of cell seeding and ECFC survival in vitro. We observed increases in mRNA expression and secretion of potent angiogenic factors by MSCs when cultured on HA-PLG scaffolds compared to PLG controls. Upon implantation into an orthotopic calvarial defect, ECFC survival on composite scaffolds was not increased in the presence of MSCs, nor did the addition of ECFCs enhance vascularization beyond increases observed with MSCs alone. Microcomputed tomography (micro-CT) performed on explanted calvarial tissues after 12 weeks revealed no significant differences between treatment groups for bone volume fraction (BVF) or bone mineral density (BMD). Taken together, these results provide evidence that HA-containing composite scaffolds seeded with MSCs can enhance neovascularization, yet MSC-secreted trophic factors do not consistently increase the persistence of co-transplanted ECFCs.


Assuntos
Cerâmica/farmacologia , Células Endoteliais/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Células-Tronco Mesenquimais/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Animais , Densidade Óssea/efeitos dos fármacos , Linhagem Celular , Técnicas de Cocultura , Ensaio de Unidades Formadoras de Colônias , DNA/metabolismo , Durapatita/farmacologia , Células Endoteliais/efeitos dos fármacos , Humanos , Ácido Láctico/farmacologia , Medições Luminescentes , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Neovascularização Fisiológica/genética , Osteogênese/efeitos dos fármacos , Ácido Poliglicólico/farmacologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Ratos , Ratos Nus , Crânio/efeitos dos fármacos , Crânio/patologia , Alicerces Teciduais/química , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Microtomografia por Raio-X
8.
Acta Biomater ; 8(2): 744-52, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22079209

RESUMO

The coating of synthetic biomaterials with cell-derived decellularized extracellular matrices (DMs) represents a promising approach to confer bioactivity to otherwise inert materials and direct cell fate of host or transplanted cells. These coatings are typically deposited on biomaterials by culturing matrix-depositing cells for a sufficient duration on the target, followed by decellularization of the substrate. We hypothesized that DMs created in monolayer culture could be collected and then transferred to a secondary substrate while retaining their instructive potential. Transferred decellularized matrices (tDMs) were created by culturing human mesenchymal stem cells (hMSCs) on tissue culture plastic (TCP) under a controlled microenvironment to deposit a highly osteogenic DM, followed by collection, mechanical homogenization and transfer to a secondary culture surface. We then investigated its capacity to accelerate naïve hMSC osteogenic differentiation by quantifying gene expression, intracellular alkaline phosphatase production, and calcium deposition when cultured on DMs or tDMs. All markers were significantly higher in hMSCs seeded on DMs or tDMs compared to cells on TCP. The osteogenic response of naïve hMSCs to tDMs was dose dependent. We observed a reduction in ERK phosphorylation in hMSCs, as well as a possible role of the cell surface integrin α2ß1, when probing the mode of efficacy for tDMs. This study represents a proof-of-principle that cell-derived matrix coatings can be deposited and effectively transferred while retaining the ability to instruct cell phenotype, thus offering a novel approach toward the development of hybrid biomaterials that mimic the complex interactions between cells and the extracellular matrix.


Assuntos
Diferenciação Celular , Matriz Extracelular/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Osteogênese , Fosfatase Alcalina/metabolismo , Antraquinonas/metabolismo , Adesão Celular , Forma Celular , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica , Humanos , Imuno-Histoquímica , Integrinas/metabolismo , Espaço Intracelular/enzimologia , Células-Tronco Mesenquimais/enzimologia , Fosforilação , Reação em Cadeia da Polimerase , Transdução de Sinais
9.
Ann Biomed Eng ; 39(4): 1174-85, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21120695

RESUMO

The presentation of extracellular matrix (ECM) proteins provides an opportunity to instruct the phenotype and behavior of responsive cells. Decellularized cell-secreted matrix coatings (DM) represent a biomimetic culture surface that retains the complexity of the natural ECM. Microenvironmental culture conditions alter the composition of these matrices and ultimately the ability of DMs to direct cell fate. We employed a design of experiments (DOE) multivariable analysis approach to determine the effects and interactions of four variables (culture duration, cell seeding density, oxygen tension, and media supplementation) on the capacity of DMs to direct the osteogenic differentiation of human mesenchymal stem cells (hMSCs). DOE analysis revealed that matrices created with extended culture duration, ascorbate-2-phosphate supplementation, and in ambient oxygen tension exhibited significant correlations with enhanced hMSC differentiation. We validated the DOE model results using DMs predicted to have superior (DM1) or lesser (DM2) osteogenic potential for naïve hMSCs. Compared to cells on DM2, hMSCs cultured on DM1 expressed 2-fold higher osterix levels and deposited 3-fold more calcium over 3 weeks. Cells on DM1 coatings also exhibited greater proliferation and viability compared to DM2-coated substrates. This study demonstrates that DOE-based analysis is a powerful tool for optimizing engineered systems by identifying significant variables that have the greatest contribution to the target output.


Assuntos
Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Osteogênese/fisiologia , Fosfatase Alcalina/metabolismo , Engenharia Biomédica , Materiais Biomiméticos , Cálcio/metabolismo , Adesão Celular , Diferenciação Celular/fisiologia , Proliferação de Células , Sobrevivência Celular , Proteínas da Matriz Extracelular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Modelos Biológicos , Osteoblastos/citologia , Osteoblastos/fisiologia , Osteogênese/genética , Reação em Cadeia da Polimerase , Engenharia Tecidual
10.
J Mol Neurosci ; 40(3): 360-6, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20107925

RESUMO

Cell-based approaches are a promising therapeutic strategy for treating injuries to the nervous system, but the optimal means to promote neurite extension and direct cellular behavior are unclear. Previous studies have examined the behavior of neural-like cells in ambient air (21% oxygen tension), yet these conditions are not representative of the physiological oxygen microenvironment of neural tissues. We hypothesized that neuronal differentiation of a model neural cell line (PC12) could be controlled by modulating local oxygen tension. Compared to ambient conditions, PC12 cells cultured in reduced oxygen exhibited significant increases in neurite extension and total neurite length, with 4% oxygen yielding the highest levels of both indicators. We confirmed neurite extension was mediated through oxygen-responsive mechanisms using small molecules that promote or inhibit HIF-1alpha stabilization. The hypoxic target gene Vegf was implicated as a neurotrophic factor, as neurite formation at 21% oxygen was mimicked with exogenous VEGF, and a VEGF-neutralizing antibody attenuated neurite formation under reduced oxygen conditions. These findings demonstrate that behavior of neural-like cells is driven by the oxygen microenvironment via VEGF function, and suggest promising approaches for future applications in neural repair.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neuritos/metabolismo , Oxigênio/metabolismo , Células PC12 , Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Diferenciação Celular/fisiologia , Humanos , Hipóxia/metabolismo , Neuritos/ultraestrutura , Células PC12/citologia , Células PC12/metabolismo , Ratos , Transdução de Sinais/fisiologia
11.
Angiogenesis ; 12(4): 303-11, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19544080

RESUMO

Therapeutic angiogenesis is a promising strategy to promote the formation of new or collateral vessels for tissue regeneration and repair. Since changes in tissue oxygen concentrations are known to stimulate numerous cell functions, these studies have focused on the oxygen microenvironment and its role on the angiogenic potential of endothelial cells. We analyzed the proangiogenic potential of human endothelial colony-forming cells (hECFCs), a highly proliferative population of circulating endothelial progenitor cells, and compared outcomes to human dermal microvascular cells (HMVECs) under oxygen tensions ranging from 1% to 21% O2, representative of ischemic or healthy tissues and standard culture conditions. Compared to HMVECs, hECFCs (1) exhibited significantly greater proliferation in both ischemic conditions and ambient air; (2) demonstrated increased migration compared to HMVECs when exposed to chemotactic gradients in reduced oxygen; and (3) exhibited comparable or superior proangiogenic potential in reduced oxygen conditions when assessed using a vessel-forming assay. These data demonstrate that the angiogenic potential of both endothelial populations is influenced by the local oxygen microenvironment. However, hECFCs exhibit a robust angiogenic potential in oxygen conditions representative of physiologic, ischemic, or ambient air conditions, and these findings suggest that hECFCs may be a superior cell source for use in cell-based approaches for the neovascularization of ischemic or engineered tissues.


Assuntos
Células Endoteliais/citologia , Hemangioblastos/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Oxigênio/farmacologia , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , Células Endoteliais/metabolismo , Sangue Fetal/citologia , Hemangioblastos/citologia , Hemangioblastos/metabolismo , Humanos , Lipoproteínas LDL/metabolismo , Veias Umbilicais/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA