Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Microcirculation ; 17(5): 367-80, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20618694

RESUMO

OBJECTIVE: Pericytes are critical cellular components of the microvasculature that play a major role in vascular development and pathologies, yet their study has been hindered by lack of a standardized method for their isolation and growth. Here we report a method for culturing human pericytes from a readily available tissue source, placenta, and provide a thorough characterization of resultant cell populations. METHODS: We developed an optimized protocol for obtaining pericytes by outgrowth from microvessel fragments recovered after enzymatic digestion of human placental tissue. We characterized outgrowth populations by immunostaining, by gene expression analysis, and by functional evaluation of cells implanted in vivo. RESULTS: Our approach yields human pericytes that may be serially expanded in culture and that uniformly express the cellular markers NG2, CD90, CD146, alpha-SMA, and PDGFR-beta, but lack markers of smooth muscle cells, endothelial cells, and leukocytes. When co-implanted with human endothelial cells into C.B-17 SCID/bg mice, human pericytes invest and stabilize developing human endothelial cell-lined microvessels. CONCLUSIONS: We conclude that our method for culturing pericytes from human placenta results in the expansion of functional pericytes that may be used to study a variety of questions related to vascular biology.


Assuntos
Técnicas de Cultura de Células/métodos , Pericitos/citologia , Pericitos/fisiologia , Animais , Biomarcadores/metabolismo , Permeabilidade Capilar , Separação Celular , Células Endoteliais/transplante , Feminino , Perfilação da Expressão Gênica , Humanos , Imunofenotipagem , Camundongos , Camundongos SCID , Microvasos/citologia , Miócitos de Músculo Liso/citologia , Análise de Sequência com Séries de Oligonucleotídeos , Pericitos/transplante , Placenta/irrigação sanguínea , Placenta/citologia , Gravidez , Transplante Heterólogo
2.
PLoS One ; 5(4): e9987, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20376322

RESUMO

BACKGROUND: Widely accessible small animal models suitable for the study of hepatitis C virus (HCV) in vivo are lacking, primarily because rodent hepatocytes cannot be productively infected and because human hepatocytes are not easily engrafted in immunodeficient mice. METHODOLOGY/PRINCIPAL FINDINGS: We report here on a novel approach for human hepatocyte engraftment that involves subcutaneous implantation of primary human fetal hepatoblasts (HFH) within a vascularized rat collagen type I/human fibronectin (rCI/hFN) gel containing Bcl-2-transduced human umbilical vein endothelial cells (Bcl-2-HUVEC) in severe combined immunodeficient X beige (SCID/bg) mice. Maturing hepatic epithelial cells in HFH/Bcl-2-HUVEC co-implants displayed endocytotic activity at the basolateral surface, canalicular microvilli and apical tight junctions between adjacent cells assessed by transmission electron microscopy. Some primary HFH, but not Huh-7.5 hepatoma cells, appeared to differentiate towards a cholangiocyte lineage within the gels, based on histological appearance and cytokeratin 7 (CK7) mRNA and protein expression. Levels of human albumin and hepatic nuclear factor 4alpha (HNF4alpha) mRNA expression in gel implants and plasma human albumin levels in mice engrafted with HFH and Bcl-2-HUVEC were somewhat enhanced by including murine liver-like basement membrane (mLBM) components and/or hepatocyte growth factor (HGF)-HUVEC within the gel matrix. Following ex vivo viral adsorption, both HFH/Bcl-2-HUVEC and Huh-7.5/Bcl-2-HUVEC co-implants sustained HCV Jc1 infection for at least 2 weeks in vivo, based on qRT-PCR and immunoelectron microscopic (IEM) analyses of gel tissue. CONCLUSION/SIGNIFICANCE: The system described here thus provides the basis for a simple and robust small animal model of HFH engraftment that is applicable to the study of HCV infections in vivo.


Assuntos
Transplante de Células/métodos , Modelos Animais de Doenças , Hepacivirus , Hepatite C , Hepatócitos/virologia , Animais , Sobrevivência Celular , Técnicas de Cocultura , Colágeno , Células Endoteliais/transplante , Células Endoteliais/virologia , Fibronectinas , Géis/química , Hepatócitos/citologia , Hepatócitos/transplante , Humanos , Camundongos , Camundongos SCID , Proteínas Proto-Oncogênicas c-bcl-2/genética , Ratos , Transplante Heterólogo , Veias Umbilicais/citologia
3.
Biomaterials ; 31(11): 3054-62, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20110124

RESUMO

Transplantation of endothelial cells (EC) for therapeutic vascularization is a promising approach in tissue engineering but has yet to be proven effective in clinical trials. This cell-based therapy is hindered by significant apoptosis of EC upon transplantation as well as poor recruitment of host mural cells to stabilize nascent vessels. Here, we address these deficiencies by augmenting endothelial cell transplantation with dual delivery of vascular endothelial growth factor (VEGF) - to improve survival of transplanted EC - and monocyte chemotactic protein-1 (MCP-1) - to induce mural cell recruitment. We produced alginate microparticles that deliver VEGF and MCP-1 with distinct release kinetics and that can be integrated into a collagen/fibronectin (protein) gel construct for delivery of EC. Combined delivery of VEGF and MCP-1 increased functional vessel formation from transplanted EC and also led to a higher number of smooth muscle cell-invested vessels than did EC therapy alone. Despite the well-known role of MCP-1 in inflammation, these beneficial effects were accomplished without a long-term increase in monocyte/macrophage recruitment or a shift to a pro-inflammatory (M1) macrophage phenotype. Overall, these data suggest a potential benefit of combined delivery of MCP-1 and VEGF from EC-containing hydrogels as a strategy for therapeutic vascularization.


Assuntos
Quimiocina CCL2 , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/transplante , Neovascularização Fisiológica/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular , Alginatos/química , Alginatos/metabolismo , Animais , Vasos Sanguíneos/citologia , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Quimiocina CCL2/administração & dosagem , Quimiocina CCL2/farmacologia , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Humanos , Hidrogéis/química , Hidrogéis/metabolismo , Implantes Experimentais , Camundongos , Microesferas , Fator A de Crescimento do Endotélio Vascular/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/farmacologia
4.
Transplantation ; 87(2): 189-97, 2009 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-19155972

RESUMO

BACKGROUND: Nearly half of all infiltrating leukocytes in rejecting human allografts are macrophages, yet, in comparison with T cells, much less is known about the contribution of this cell type to rejection. Our laboratory has previously described models of rejection of human skin or artery grafts in immunodeficient mouse hosts mediated by adoptively transferred allogeneic T cells. However, mature human monocyte/macrophages have consistently failed to engraft in these animals. Here, we describe the introduction of human CD68+ macrophages into irradiated immunodeficient mice by transplantation of enriched CD34+ hematopoietic stem-cells isolated from peripheral blood of G-colony-stimulating factor pretreated adults. METHODS: We investigated strains of immunodeficient mice bearing human tissue grafts (skin and artery) inoculated with 1 x 10(6) human CD34+ adult hematopoietic stem cells, peripheral blood monuclear cells autologous to the CD34 donor, or both for human cell engraftment. RESULTS: In the absence of T cells, CD68+ CD14+ macrophages infiltrate allogeneic human skin but produce little injury or thrombosis. Both responses are enhanced when combined with adoptive transfer of T cells autologous to the hematopoietic stem cells as exemplified by the induction of the macrophage activation marker CD163. CD68+ macrophages also infiltrate allogeneic arterial interposition grafts, producing intimal expansion and calcification in the absence of T cells. CONCLUSIONS: These new models may be used to study the role of human macrophages in transplant rejection and other pathologies in vivo.


Assuntos
Células-Tronco Adultas/imunologia , Artérias/imunologia , Artérias/transplante , Rejeição de Enxerto/imunologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/imunologia , Macrófagos/imunologia , Transplante de Pele/imunologia , Pele/imunologia , Transferência Adotiva , Animais , Antígenos CD/análise , Antígenos CD34/análise , Antígenos de Diferenciação Mielomonocítica/análise , Artérias/patologia , Modelos Animais de Doenças , Rejeição de Enxerto/patologia , Humanos , Subunidade gama Comum de Receptores de Interleucina/genética , Subunidade gama Comum de Receptores de Interleucina/imunologia , Receptores de Lipopolissacarídeos/análise , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Receptores de Superfície Celular/análise , Pele/patologia , Linfócitos T/imunologia , Linfócitos T/transplante , Transplante Homólogo , Irradiação Corporal Total
5.
Tissue Eng Part A ; 15(1): 165-73, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18620481

RESUMO

Collagen-fibronectin gels containing Bcl-2-transduced human umbilical vein endothelial cells (Bcl-2-HUVEC) implanted in the abdominal walls of immunodeficient mice form mature microvessels invested by host-derived smooth muscle cells (SMC) by 8 weeks. We tested the hypothesis that coengraftment of human aortic SMC (HASMC) could accelerate vessel maturation. To prevent SMC-mediated gel contraction, we polymerized the gel within a nonwoven poly(glycolic acid) (PGA) scaffold. Implanted grafts were evaluated at 15, 30, and 60 days. Acellular PGA-supported protein gels elicited a macrophage-rich foreign body reaction and transient host angiogenic response. When transplanted alone, HASMC tightly associated with the fibers of the scaffold and incorporated into the walls of angiogenic mouse microvessels, preventing their regression. When transplanted alone in PGA-supported gels, Bcl-2-HUVEC retained the ability to form microvessels invested by mouse SMC. Interestingly, grafts containing both Bcl-2-HUVEC and HASMC displayed greater numbers of smooth muscle alpha-actin-expressing cells associated with human EC-lined arteriole-like microvessels at all times examined and showed a significant increase in the number of larger caliber microvessels at 60 days. We conclude that SMC coengraftment can accelerate vessel development by EC and promote arteriolization. This strategy of EC-SMC coengraftment in PGA-supported protein gels may have broader application for perfusing bioengineered tissues.


Assuntos
Arteríolas/citologia , Células Endoteliais/transplante , Endotélio Vascular/citologia , Miócitos de Músculo Liso/citologia , Neovascularização Fisiológica , Animais , Aorta/citologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Humanos , Imuno-Histoquímica , Camundongos , Camundongos SCID , Ácido Poliglicólico/química , Ácido Poliglicólico/farmacologia , Fatores de Tempo , Engenharia Tecidual , Alicerces Teciduais , Transplante Heterólogo , Veias Umbilicais/citologia
6.
FASEB J ; 22(8): 2949-56, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18450813

RESUMO

Transplantation of Bcl-2-transduced human umbilical vein endothelial cells (ECs) in protein gels into the gastrocnemius muscle improves local reperfusion in immunodeficient mouse hosts with induced hind limb ischemia. We tested the hypothesis that incorporation of local, sustained growth factor delivery could enhance and accelerate this effect. Tissue engineering scaffolds often use synthetic polymers to enable controlled release of proteins, but most synthetic delivery systems have major limitations, most notably hydrophobicity and inefficient protein loading. Here, we report the development of a novel alginate-based delivery system for vascular endothelial growth factor-A(165) (VEGF) that exhibits superior loading efficiency and physical properties to previous systems in vitro. In vivo, VEGF released from alginate microparticles within protein gels was biologically active and, when combined with EC transplantation, led to increased survival of transplanted cells at 28 days. The composite graft described also improved early (14 days) tissue perfusion and late (28 days) muscle myoglobin expression, a sign of recovery from ischemia, compared with EC transplantation and VEGF delivery separately. We conclude that our improved approach to sustained VEGF delivery in tissue engineering is useful in vivo and that the integration of high efficiency protein delivery enhances the therapeutic effect of protein gel-based EC transplantation.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Células Endoteliais/transplante , Engenharia Tecidual/métodos , Fator A de Crescimento do Endotélio Vascular/administração & dosagem , Alginatos , Animais , Bovinos , Sobrevivência Celular , Células Cultivadas , Colágeno/administração & dosagem , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Fibronectinas/administração & dosagem , Géis , Genes bcl-2 , Ácido Glucurônico , Ácidos Hexurônicos , Membro Posterior/irrigação sanguínea , Humanos , Isquemia/patologia , Isquemia/fisiopatologia , Isquemia/terapia , Ácido Láctico , Camundongos , Camundongos Endogâmicos C57BL , Microesferas , Tamanho da Partícula , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Soroalbumina Bovina/administração & dosagem , Transdução Genética
7.
J Cell Physiol ; 216(2): 389-95, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18247368

RESUMO

Vascular smooth muscle cells (SMC) may be directly exposed to blood flow after an endothelial-denuding injury. It is not known whether direct exposure of SMC to shear stress reduces SMC turnover and contributes to the low rate of restenosis after most vascular interventions. This study examines if laminar shear stress inhibits SMC proliferation or stimulates apoptosis. Bovine aortic SMC were exposed to arterial magnitudes of laminar shear stress (11 dynes/cm(2)) for up to 24 h and compared to control SMC (0 dynes/cm(2)). SMC density was assessed by cell counting, DNA synthesis by (3)[H]-thymidine incorporation, and apoptosis by TUNEL staining. Akt, caspase, bax, and bcl-2 phosphorylation were assessed by Western blotting; caspase activity was also measured with an in vitro assay. Analysis of variance was used to compare groups. SMC exposed to laminar shear stress had a 38% decrease in cell number (n = 4, P = 0.03), 54% reduction in (3)[H]-thymidine incorporation (n = 3, P = 0.003), and 15-fold increase in TUNEL staining (n = 4, P < 0.0001). Akt phosphorylation was reduced by 67% (n = 3, P < 0.0001), whereas bax/bcl-2 phosphorylation was increased by 1.8-fold (n = 3, P = 0.01). Caspase-3 activity was increased threefold (n = 5, P = 0.03). Pretreatment of cells with ZVAD-fmk or wortmannin resulted in 42% increased cell retention (n = 3, P < 0.01) and a fourfold increase in apoptosis (n = 3, P < 0.04), respectively. Cells transduced with constitutively-active Akt had twofold decreased apoptosis (n = 3, P < 0.002). SMC exposed to laminar shear stress have decreased proliferation and increased apoptosis, mediated by the Akt pathway. These results suggest that augmentation of SMC apoptosis may be an alternative strategy to inhibit restenosis after vascular injury.


Assuntos
Apoptose/fisiologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia , Animais , Bovinos , Proliferação de Células , Células Cultivadas , Hemodinâmica , Humanos , Marcação In Situ das Extremidades Cortadas , Miócitos de Músculo Liso/citologia , Proteínas Proto-Oncogênicas c-akt/genética , Resistência ao Cisalhamento , Estresse Mecânico
8.
J Cell Biol ; 180(1): 101-12, 2008 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-18195103

RESUMO

Prohibitin 1 (PHB1) is a highly conserved protein that is mainly localized to the inner mitochondrial membrane and has been implicated in regulating mitochondrial function in yeast. Because mitochondria are emerging as an important regulator of vascular homeostasis, we examined PHB1 function in endothelial cells. PHB1 is highly expressed in the vascular system and knockdown of PHB1 in endothelial cells increases mitochondrial production of reactive oxygen species via inhibition of complex I, which results in cellular senescence. As a direct consequence, both Akt and Rac1 are hyperactivated, leading to cytoskeletal rearrangements and decreased endothelial cell motility, e.g., migration and tube formation. This is also reflected in an in vivo angiogenesis assay, where silencing of PHB1 blocks the formation of functional blood vessels. Collectively, our results provide evidence that PHB1 is important for mitochondrial function and prevents reactive oxygen species-induced senescence and thereby maintains the angiogenic capacity of endothelial cells.


Assuntos
Senescência Celular , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Mitocôndrias/fisiologia , Neovascularização Fisiológica , Proteínas Repressoras/fisiologia , Animais , Aorta/citologia , Aorta/metabolismo , Células Cultivadas , Citoesqueleto/metabolismo , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/metabolismo , Células Endoteliais/citologia , Endotélio Vascular/citologia , Humanos , Camundongos , Camundongos Endogâmicos , Mitocôndrias/metabolismo , Membranas Mitocondriais/metabolismo , Neovascularização Fisiológica/genética , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proibitinas , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais , Proteínas rac de Ligação ao GTP/genética , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP
9.
J Immunol ; 179(11): 7488-96, 2007 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18025193

RESUMO

There is considerable interest in exploiting circulating endothelial progenitor cells (EPCs) for therapeutic organ repair. Such cells may be differentiated into endothelial cells (ECs) in vitro and then expanded for use in tissue engineering. Vessel-derived ECs are variably immunogenic, depending upon tissue source, and it is unknown whether ECs derived from cord blood EPCs are able to initiate an allogeneic response. In this study, we compare the phenotype and alloantigenicity of human cord blood progenitor cell-derived ECs with HUVECs isolated from the same donors. Human cord blood progenitor cell-derived ECs are very similar to HUVECs in the expression of proteins relevant for alloimmunity, including MHC molecules, costimulators, adhesion molecules, cytokines, chemokines, and IDO, and in their ability to initiate allogeneic CD4(+) and CD8(+) memory T cell responses in vitro and in vivo. These findings have significant implications for the use of cord blood EPCs in regenerative medicine or tissue engineering.


Assuntos
Células Endoteliais/imunologia , Sangue Fetal/imunologia , Células-Tronco Hematopoéticas/imunologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Células Cultivadas , Células Endoteliais/citologia , Sangue Fetal/citologia , Células-Tronco Hematopoéticas/citologia , Humanos , Imunidade , Memória Imunológica/imunologia , Fenótipo , Fatores de Tempo
10.
J Immunol ; 179(7): 4397-404, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17878335

RESUMO

The frequency of circulating alloreactive human memory T cells correlates with allograft rejection. Memory T cells may be divided into effector memory (T(EM)) and central memory (T(CM)) cell subsets, but their specific roles in allograft rejection are unknown. We report that CD4+ T(EM) (CD45RO+ CCR7- CD62L-) can be adoptively transferred readily into C.B-17 SCID/bg mice and mediate the destruction of human endothelial cells (EC) in vascularized human skin grafts allogeneic to the T cell donor. In contrast, CD4+ T(CM) (CD45RO+ CCR7+ CD62L+) are inefficiently transferred and do not mediate EC injury. In vitro, CD4+ T(EM) secrete more IFN-gamma within 48 h in response to allogeneic ECs than do T(CM). In contrast, T(EM) and T(CM) secrete comparable amounts of IFN-gamma in response to allogeneic monocytes (Mo). In the same cultures, both T(EM) and T(CM) produce IL-2 and proliferate in response to IFN-gamma-treated allogeneic human EC or Mo, but T(CM) respond more vigorously in both assays. Blockade of LFA-3 strongly inhibits both IL-2 and IFN-gamma secretion by CD4+ T(EM) cultured with allogeneic EC but only minimally inhibits responses to allogeneic Mo. Blockade of CD80 and CD86 strongly inhibits IL-2 but not IFN-gamma production by in response to allogeneic EC or Mo. Transduction of EC to express B7-2 enhances allogeneic T(EM) production of IL-2 but not IFN-gamma. We conclude that human CD4+ T(EM) directly recognize and respond to allogeneic EC in vitro by secreting IFN-gamma and that this response depends on CD2 but not CD28. Consistent with EC activation of effector functions, human CD4+ T(EM) can mediate allogeneic EC injury in vivo.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Células Endoteliais/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Antígeno B7-1/imunologia , Linfócitos T CD4-Positivos/metabolismo , Antígenos CD58/imunologia , Células Cultivadas , Feminino , Humanos , Memória Imunológica/imunologia , Interferon gama/biossíntese , Interleucina-2/biossíntese , Camundongos , Camundongos SCID
11.
Tissue Eng ; 13(12): 2871-9, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17883324

RESUMO

The primary objective of this study was to evaluate epicardial transplantation of an intact microvascular network for treatment of myocardial ischemia in a murine model of acute myocardial infarction. We describe transplantation of an intact microvascular network constructed from isolated microvascular segments stabilized in a 3-dimensional matrix to the epicardial surface after acute myocardial infarction. This microvascular graft was implanted as a patch on the epicardium of mice after left coronary artery ligation. After 14 and 28 days of implantation, left ventricular (LV) function was assessed and grafts evaluated via histology and cytochemistry. Inosculation of microvessels within the graft with host coronary microcirculation occurred as early as 7 days after initial tissue grafting. Morphologic evaluation of the grafts revealed arterioles, venules, capillaries, and erythrocytes within vascular lumina. Control grafts of collagen alone remained avascular. LV infarct size was smaller, and LV function improved in treated animals. Engraftment of whole microvascular units can be achieved to support cell-assisted vascular remodeling. Microvascular grafts may provide therapeutic benefit as a primary treatment or serve as a microvascular platform for cardiac repair and regeneration.


Assuntos
Prótese Vascular , Capilares/patologia , Capilares/transplante , Infarto do Miocárdio/patologia , Infarto do Miocárdio/cirurgia , Animais , Análise de Falha de Equipamento , Camundongos , Ratos , Ratos Endogâmicos F344 , Resultado do Tratamento
12.
FASEB J ; 20(10): 1739-41, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16807367

RESUMO

We seeded tissue engineered human skin substitutes with endothelial cells (EC) differentiated in vitro from progenitors from umbilical cord blood (CB-EC) or adult peripheral blood (AB-EC), comparing the results to previous work using cultured human umbilical vein EC (HUVEC) with or without Bcl-2 transduction. Vascularized skin substitutes were prepared by seeding Bcl-2-transduced or nontransduced HUVEC, CB-EC, or AB-EC on the deep surface of decellularized human dermis following keratinocyte coverage of the epidermal surface. These skin substitutes were transplanted onto C.B-17 SCID/beige mice receiving systemic rapamycin or vehicle control and were analyzed 21 d later. CB-EC and Bcl-2-HUVEC formed more human EC-lined vessels than AB-EC or control HUVEC; CB-EC, Bcl-2-HUVEC, and AB-EC but not control HUVEC promoted ingrowth of mouse EC-lined vessels. Bcl-2 transduction increased the number of human and mouse EC-lined vessels in grafts seeded with HUVEC but not with CB-EC or AB-EC. Both CB-EC and AB-EC-induced microvessels became invested by smooth muscle cell-specific alpha-actin-positive mural cells, indicative of maturation. Rapamycin inhibited ingrowth of mouse EC-lined vessels but did not inhibit formation of human EC-lined vessels. We conclude that EC differentiated from circulating progenitors can be utilized to vascularize human skin substitutes even in the setting of compromised host angiogenesis/vasculogenesis.


Assuntos
Neovascularização Fisiológica , Pele Artificial , Engenharia Tecidual/métodos , Animais , Células Endoteliais/citologia , Endotélio Vascular/citologia , Humanos , Camundongos , Camundongos SCID , Proteínas Proto-Oncogênicas c-bcl-2/genética , Sirolimo/farmacologia , Células-Tronco/citologia , Transdução Genética , Transplante Heterólogo
13.
Tissue Eng ; 11(11-12): 1678-87, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16411813

RESUMO

The current experiments used a scaffold-based, three-dimensional, human dermal fibroblast culture (3DFC) as a cardiac patch to stimulate revascularization and preserve left ventricular (LV) function of the infarcted LV in severe combined immunodeficient (SCID) mice. The 3DFC contains viable cells that secrete angiogenic growth factors and has been previously shown to stimulate angiogenesis. The hypothesis tested was that a 3DFC cardiac patch would attenuate a reduction in LV function of infarcted hearts. Five groups of mice were studied, including normal SCID mice (n = 13), normal SCID mice with 3DFC (n = 6), infarcted SCID mice (n = 6), infarcted mice with nonviable 3DFC (n = 6), and infarcted SCID mice with 3DFC (n = 6). An occlusion of a branch of the left anterior descending (LAD) coronary artery was performed by thermal ligation, and 3DFC was sized to the damaged area and implanted onto the epicardium at the site of tissue injury. Fourteen days postsurgery, LV mechanics were characterized with the Millar conductance catheter system (CCS). The data demonstrated that 3DFC-treated infarcted myocardium had significantly higher ejection fractions (EFs) compared with infarct-only mice (58.9 +/- 10.8 versus 31.0 +/- 5.8%, respectively; p < 0.05). Preload recruitable stroke work (PRSW) parameters were significantly higher in 3DFC-treated mice compared with infarct-only mice (64.6 +/- 11.9 versus 36.8 +/- 6.4 mmHg, respectively; p < 0.05). These results show that the 3DFC as a cardiac patch functioned to attenuate further loss of LV function accompanying acute myocardial infarct and that this may be related in part to myocardial revascularization.


Assuntos
Fibroblastos/transplante , Infarto do Miocárdio/terapia , Revascularização Miocárdica , Recuperação de Função Fisiológica , Função Ventricular Esquerda , Animais , Células Cultivadas , Feminino , Fibroblastos/metabolismo , Humanos , Camundongos , Camundongos SCID , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Revascularização Miocárdica/métodos
14.
Proc Natl Acad Sci U S A ; 102(2): 425-30, 2005 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-15625106

RESUMO

Implants of collagen-fibronectin gels containing Bcl-2-transduced human umbilical vein endothelial cells (Bcl-2-HUVECs) induce the formation of human endothelial cell (EC)/murine vascular smooth muscle cell (VSMC) chimeric vessels in immunodeficient mice. Microfil casting of the vasculature 60 d after implantation reveals highly branched microvascular networks within the implants that connect with and induce remodeling of conduit vessels arising from the abdominal wall circulation. Approximately 85% of vessels within the implants are lined by Bcl-2-positive human ECs expressing VEGFR1, VEGFR2, and Tie-2, but not integrin alpha(v)beta(3). The human ECs are seated on a well formed human laminin/collagen IV-positive basement membrane, and are surrounded by mouse VSMCs expressing SM-alpha actin, SM myosin, SM22alpha, and calponin, all markers of contractile function. Transmission electron microscopy identified well formed EC-EC junctions, chimeric arterioles with concentric layers of contractile VSMC, chimeric capillaries surrounded by pericytes, and chimeric venules. Bcl-2-HUVEC-lined vessels retain 70-kDa FITC-dextran, but not 3-kDa dextran; local histamine rapidly induces leak of 70-kDa FITC-dextran or India ink. As in skin, TNF induces E-selectin and vascular cell adhesion molecule 1 only on venular ECs, whereas intercellular adhesion molecule-1 is up-regulated on all human ECs. Bcl-2-HUVEC implants are able to engraft within and increase perfusion of ischemic mouse gastrocnemius muscle after femoral artery ligation. These studies show that cultured Bcl-2-HUVECs can differentiate into arterial, venular, and capillary-like ECs when implanted in vivo, and induce arteriogenic remodeling of the local mouse vessels. Our results support the utility of differentiated EC transplantation to treat tissue ischemia.


Assuntos
Células Endoteliais/transplante , Isquemia/terapia , Neovascularização Fisiológica , Proteínas Proto-Oncogênicas c-bcl-2/genética , Animais , Permeabilidade Capilar , Diferenciação Celular , Membro Posterior/irrigação sanguínea , Histamina/farmacologia , Humanos , Imuno-Histoquímica , Camundongos , Microscopia Eletrônica , Transdução Genética , Fator de Necrose Tumoral alfa/farmacologia
15.
Arterioscler Thromb Vasc Biol ; 24(5): 898-904, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-14988090

RESUMO

OBJECTIVE: We have previously demonstrated the ability to construct 3-dimensional microvascular beds in vitro via angiogenesis from isolated, intact, microvessel fragments that retain endothelial cells and perivascular cells. Our objective was to develop and characterize an experimental model of tissue vascularization, based on the implantation of this microvascular construct, which recapitulated angiogenesis, vessel differentiation, and network maturation. METHODS AND RESULTS: On implantation in a severe combined-immunodeficient mouse model, vessels in the microvascular constructs rapidly inosculated with the recipient host circulation. Ink perfusion of implants via the left ventricle of the host demonstrated that vessel inosculation begins within the first day after implantation. Evaluation of explanted constructs over the course of 28 days revealed the presence of a mature functional microvascular bed. Using a probe specific for the original microvessel source, 91.7%+/-11% and 88.6%+/-19% of the vessels by day 5 and day 28 after implantation, respectively, were derived from the original microvessel isolate. Similar results were obtained when human-derived microvessels were used to build the microvascular construct. CONCLUSIONS: With this model, we reproduce the important aspects of vascularization, angiogenesis, inosculation, and network remodeling. Furthermore, we demonstrate that the model accommodates human-derived vessel fragments, enabling the construction of human-mouse vascular chimeras.


Assuntos
Vasos Sanguíneos/transplante , Microcirculação/fisiologia , Neovascularização Fisiológica , Animais , Carbono , Colágeno , Corantes , Epididimo , Feminino , Géis , Humanos , Masculino , Camundongos , Camundongos SCID , Morfogênese , Músculo Liso Vascular/citologia , Técnicas de Cultura de Órgãos/métodos , Ratos , Ratos Sprague-Dawley , Tela Subcutânea
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA