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










Base de dados
Intervalo de ano de publicação
1.
J Thromb Haemost ; 22(3): 715-726, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37940047

RESUMO

BACKGROUND: Fibrinogen is a plasma protein forming the fibrin scaffold of blood clots. Its mechanical properties therefore affect the risk of bleeding as well as thrombosis. There has been much recent interest in the biophysical mechanisms controlling fibrin mechanics; however, the role of molecular heterogeneity of the circulating fibrinogen in determining clot mechanical function remains poorly characterized. OBJECTIVES: By comparing 2 fibrinogen variants where the only difference is the Aα-chain length, with one variant having a globular domain at its C-terminus, this study aimed to reveal how the molecular structure impacts the structure and mechanics of fibrin networks. METHODS: We characterized the mechanical response to large shear for networks formed from 2 recombinant fibrinogen variants: the most prevalent variant in circulation with a molecular weight of 340 kDa (recombinant human fibrinogen [rFib] 340) and a minor variant with a molecular weight of 420 kDa (rFib420). RESULTS: We show that the elastic properties of the 2 variants are identical when fibrin is cross-linked with factor XIIIa but differ strongly in its absence. Uncross-linked rFib420 networks are softer and up to 3-fold more extensible than rFib340 networks. Electron microscopy imaging showed that the 2 variants formed networks with a comparable structure, except at 4 mg/mL, where rFib420 formed denser networks. CONCLUSION: We propose that the αEC domains of rFib420 increase the extensibility of uncross-linked fibrin networks by promoting protofibril sliding, which is blocked by FXIIIa cross-linking. Our findings can help explain the functional role of different circulating fibrinogen variants in blood clot mechanics and tissue repair.


Assuntos
Hemostáticos , Trombose , Humanos , Fibrina/química , Fator XIIIa/química , Fibrinogênio/metabolismo , Coagulação Sanguínea
2.
J Thromb Haemost ; 21(8): 2277-2290, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37001817

RESUMO

BACKGROUND: Staphylococcus aureus is a common gram-positive bacterium that is the causative agent for several human diseases, including sepsis. A key virulence mechanism is pathogen binding to host fibrinogen through the C-terminal region of the γ-chain. Previous work demonstrated that FggΔ5 mice expressing mutant fibrinogen γΔ5 lacking a S. aureus binding motif had significantly improved survival following S. aureus septicemia. Fibrinogen γ' is a human splice variant that represents about 10% to 15% of the total fibrinogen in plasma and circulates as a fibrinogen γ'-γ heterodimer (phFibγ'-γ). The fibrinogen γ'-chain is also expected to lack S. aureus binding function. OBJECTIVE: Determine if human fibrinogen γ'-γ confers host protection during S. aureus septicemia. METHODS: Analyses of survival and the host response following S. aureus septicemia challenge in FggΔ5 mice and mice reconstituted with purified phFibγ'-γ or phFibγ-γ. RESULTS: Reconstitution of fibrinogen-deficient or wildtype mice with purified phFibγ'-γ prior to infection provided a significant prolongation in host survival relative to mice reconstituted with purified phFibγ-γ, which was superior to that observed with heterozygous FggΔ5 mice. Improved survival could not be accounted for by quantitative differences in fibrinogen-dependent adhesion or clumping, but phFibγ'-γ-containing mixtures generated notably smaller bacterial aggregates. Importantly, administration of phFibγ'-γ after infection also provided a therapeutic benefit by prolonging host survival relative to administration of phFibγ-γ. CONCLUSION: These findings provide the proof-of-concept that changing the ratio of naturally occurring fibrinogen variants in blood could offer significant therapeutic potential against bacterial infection and potentially other diseases.


Assuntos
Bacteriemia , Fibrinogênios Anormais , Sepse , Infecções Estafilocócicas , Humanos , Camundongos , Animais , Staphylococcus aureus/metabolismo , Fibrinogênio/metabolismo
3.
Blood Coagul Fibrinolysis ; 26(6): 613-20, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26083991

RESUMO

Fibrinogen as precursor of fibrin plays an essential role in clot formation. There are three main mechanisms associated with a reduction in fibrinogen concentration during cardiac surgery: hemodilution, consumption, and degradation. Moreover, early fibrinogen degradation products (FgDPs) can interfere with normal fibrin formation of intact fibrinogen. The aim of this study was to determine the relative contributions of hemodilution, consumption, and degradation to fibrinogen loss in cardiac surgery and to evaluate the effects fibrinogen degradation products on blood clot formation in vitro. First, fibrin and fibrinogen concentrations, their degradation products, hematocrit, and albumin concentrations were compared in 10 patients before and after isolated coronary artery bypass graft (CABG) surgery with cardiopulmonary bypass. Second, ex-vivo fibrinogen supplementation experiments were performed. Finally, the effects of purified FgDPs on clotting time and clot firmness were established in vitro in whole blood by ROTEM. Fibrinogen plasma concentration decreased 30% during surgery. This drop appears to be mainly caused by hemodilution, as both hematocrit and albumin levels decreased and no relevant increase in D-dimer levels and FgDPs was observed. Furthermore, the coagulation profile normalized after addition of purified fibrinogen. Early FgDPs demonstrated a significant impact on in-vitro whole blood clotting. Although early FgDPs have a pronounced effect on blood clot formation in vitro and therefore may induce or enhance in vivo coagulopathy, the drop of fibrinogen concentration seen after CABG surgery (using tranexamic acid) is primarily caused by hemodilution.


Assuntos
Testes de Coagulação Sanguínea/métodos , Procedimentos Cirúrgicos Cardíacos/métodos , Ponte Cardiopulmonar/métodos , Fibrinogênio/metabolismo , Feminino , Humanos , Masculino
4.
Thromb Res ; 132(1): e48-53, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23642654

RESUMO

INTRODUCTION: Fibrinogen is a key component of the blood coagulation system and plays important, diverse roles in several relevant pathologies such as thrombosis, hemorrhage, and cancer. It is a large glycoprotein whose three-dimensional molecular structure is not fully known. Furthermore, circulating fibrinogen is highly heterogeneous, mainly due to proteolytic degradation and alternative mRNA processing. Recombinant production of human fibrinogen allows investigating the impact on the three-dimensional structure of specific changes in the primary structure. METHODS: We performed analytical ultracentrifugation analyses of a full-length recombinant human fibrinogen, its counterpart purified from human plasma, and a recombinant human fibrinogen with both Aα chains truncated at amino acid 251, thus missing their last 359 amino acid residues. RESULTS: We have accurately determined the translational diffusion and sedimentation coefficients (Dt(20,w)(0), s(20,w)(0)) of all three species. This was confirmed by derived molecular weights within 1% for the full length species, and 5% for the truncated species, as assessed by comparison with SDS-PAGE/Western blot analyses and primary structure data. No significant differences in the values of Dt(20,w)(0) and s(20,w)(0) were found between the recombinant and purified full length human fibrinogens, while slightly lower and higher values, respectively, resulted for the recombinant truncated human fibrinogen compared to a previously characterized purified human fibrinogen fragment X obtained by plasmin digestion. CONCLUSIONS: Full-length recombinant fibrinogen is less polydisperse but hydrodynamically indistinguishable from its counterpart purified from human plasma. Recombinant Aα251-truncated human fibrinogen instead behaves differently from fragment X, suggesting a role for the Bß residues 1-52 in inter-molecular interactions. Overall, these new hydrodynamic data will constitute a reliable benchmark against which models of fibrinogen species could be compared.


Assuntos
Fibrinogênio/química , Western Blotting , Cromatografia de Fase Reversa , Eletroforese em Gel de Poliacrilamida , Produtos de Degradação da Fibrina e do Fibrinogênio/química , Produtos de Degradação da Fibrina e do Fibrinogênio/genética , Fibrinogênio/genética , Fibrinogênio/isolamento & purificação , Humanos , Hidrodinâmica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Ultracentrifugação
5.
Cardiovasc Res ; 88(2): 367-75, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20562095

RESUMO

AIMS: Matrix metalloproteinases (MMP) and plasminogen activator (PA)/plasmin-mediated proteolysis, especially at the cell surface, play important roles in matrix degeneration and smooth muscle cell migration, which largely contributes to vein graft failure. In this study, a novel hybrid protein was designed to inhibit both protease systems simultaneously. MMP and plasmin activity were inhibited at the cell surface by this hybrid protein, consisting of the receptor-binding amino-terminal fragment (ATF) of urokinase-type PA, linked to both the tissue inhibitor of metalloproteinases (TIMP-1) and bovine pancreas trypsin inhibitor (BPTI), a potent protease inhibitor. The effect of overexpression of this protein on vein graft disease was studied. METHODS AND RESULTS: A non-viral expression vector encoding the hybrid protein TIMP-1.ATF.BPTI was constructed and validated. Next, cultured segments of human veins were transfected with this vector. Expressing TIMP-1.ATF.BPTI in vein segments resulted in a mean 36 ± 14% reduction in neointima formation after 4 weeks. In vivo inhibition of vein graft disease by TIMP-1.ATF.BPTI is demonstrated in venous interpositions placed into carotid arteries of hypercholesterolaemic APOE*3Leiden mice. After 4 weeks, vein graft thickening was significantly inhibited in mice treated with the domains TIMP-1, ATF, or BPTI (36-49% reduction). In the TIMP-1.ATF.BPTI-treated mice, vein graft thickening was reduced by 67±4%, which was also significantly stronger when compared with the individual components. CONCLUSION: These data provide evidence that cell surface-bound inhibition of the PA and MMP system by the hybrid protein TIMP-1.ATF.BPTI, overexpressed in distant tissues after electroporation-mediated non-viral gene transfer, is a powerful approach to prevent vein graft disease.


Assuntos
Proliferação de Células , Fibrinolisina/metabolismo , Terapia Genética , Oclusão de Enxerto Vascular/prevenção & controle , Metaloproteinases da Matriz/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Veia Safena/metabolismo , Veias Cavas/metabolismo , Animais , Apolipoproteína E3/genética , Aprotinina/biossíntese , Aprotinina/genética , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Artérias Carótidas/cirurgia , Bovinos , Linhagem Celular , Modelos Animais de Doenças , Eletroporação , Fibrinolisina/antagonistas & inibidores , Terapia Genética/métodos , Oclusão de Enxerto Vascular/genética , Oclusão de Enxerto Vascular/metabolismo , Oclusão de Enxerto Vascular/patologia , Humanos , Hipercolesterolemia/genética , Hipercolesterolemia/metabolismo , Hipercolesterolemia/patologia , Hiperplasia , Masculino , Inibidores de Metaloproteinases de Matriz , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteínas Recombinantes de Fusão/biossíntese , Veia Safena/patologia , Veia Safena/cirurgia , Fatores de Tempo , Técnicas de Cultura de Tecidos , Inibidor Tecidual de Metaloproteinase-1/biossíntese , Inibidor Tecidual de Metaloproteinase-1/genética , Transfecção , Ativador de Plasminogênio Tipo Uroquinase/biossíntese , Ativador de Plasminogênio Tipo Uroquinase/genética , Veias Cavas/patologia , Veias Cavas/transplante
6.
J Vasc Surg ; 51(2): 429-37, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20036101

RESUMO

BACKGROUND: Smooth muscle cell (SMC) migration and proliferation are important in the development of intimal hyperplasia, the major cause of vein graft failure. Proteases of the plasminogen activator (PA) system and of the matrix metalloproteinase (MMP) system are pivotal in extracellular matrix degradation and, by that, SMC migration. Previously, we demonstrated that inhibition of both protease systems simultaneously with viral gene delivery of the hybrid protein TIMP-1.ATF, consisting of the tissue inhibitor of metalloproteinase-1 (TIMP-1) and the receptor-binding amino terminal fragment (ATF) of urokinase, reduces SMC migration and neointima formation in an in vitro restenosis model using human saphenous vein cultures more efficiently than both protease systems separately. Because use of viral gene delivery is difficult in clinical application, this study used nonviral delivery of TIMP-1.ATF plasmid to reduce vein graft disease in a murine bypass model. Nonviral gene transfer by electroporation was used to avert major disadvantages of viral gene delivery, such as immune responses and short-term expression. METHODS: Plasmids encoding ATF, TIMP-1, TIMP-1.ATF, or luciferase, as a control, were injected and electroporated in both calf muscles of hypercholesterolemic apolipoprotein E3-Leiden (APOE*3Leiden) mice (n = 8). One day after electroporation, a venous interposition of a donor mouse was placed into the carotid artery of a recipient mouse. In this model, vein graft thickening develops with features of accelerated atherosclerosis. Vein grafts were harvested 4 weeks after electroporation and surgery, and histologic analysis of the vessel wall was performed. RESULTS: Electroporation-mediated overexpression of the plasmid vectors resulted in a prolonged expression of the transgenes and resulted in a significant reduction of vein graft thickening (ATF: 36% +/- 9%, TIMP-1: 49% +/- 5%, TIMP-1.ATF: 58% +/- 5%; P < .025). Although all constructs reduced vein graft thickening compared with the controls, the luminal area was best preserved in the TIMP-1.ATF-treated mice. CONCLUSION: Intramuscular electroporation of TIMP-1.ATF inhibits vein graft thickening in vein grafts in carotid arteries of hypercholesterolemic mice. Binding of TIMP-1.ATF hybrid protein to the u-PA receptor at the cell surface enhances the inhibitory effect of TIMP-1 on vein graft remodeling in vitro as well as in vivo and may be an effective strategy to prevent vein graft disease.


Assuntos
Aterosclerose/prevenção & controle , Eletroporação , Técnicas de Transferência de Genes , Terapia Genética/métodos , Oclusão de Enxerto Vascular/prevenção & controle , Inibidor Tecidual de Metaloproteinase-1/biossíntese , Ativador de Plasminogênio Tipo Uroquinase/biossíntese , Veias Cavas/transplante , Animais , Apolipoproteína E3/genética , Aterosclerose/enzimologia , Aterosclerose/genética , Aterosclerose/patologia , Artérias Carótidas/cirurgia , Modelos Animais de Doenças , Oclusão de Enxerto Vascular/enzimologia , Oclusão de Enxerto Vascular/genética , Oclusão de Enxerto Vascular/patologia , Sobrevivência de Enxerto , Humanos , Hipercolesterolemia/genética , Hipercolesterolemia/patologia , Hipercolesterolemia/terapia , Hiperplasia , Masculino , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mutação , Fragmentos de Peptídeos/biossíntese , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Proteínas Recombinantes de Fusão/biossíntese , Fatores de Tempo , Inibidor Tecidual de Metaloproteinase-1/sangue , Inibidor Tecidual de Metaloproteinase-1/genética , Ativador de Plasminogênio Tipo Uroquinase/sangue , Ativador de Plasminogênio Tipo Uroquinase/genética , Veias Cavas/enzimologia , Veias Cavas/patologia
7.
Hum Gene Ther ; 18(9): 861-9, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17850190

RESUMO

For the successful application of RNA interference in vivo, it is desired to achieve (local) delivery of small interfering RNAs (siRNAs) and long-term gene silencing. Nonviral electrodelivery is suitable to obtain local and prolonged expression of transgenes. By intramuscular electrodelivery of a plasmid in which two opposing human polymerase III promoters (H1 and U6) drive the expression of siRNA constructs that form functional double-stranded siRNAs, in combination with in vivo bioluminescence imaging, we were able to knock down exogenous delivered luciferase for at least 100 days in murine calf muscles. This effect was sequence specific, because scrambled siRNA had no effect. Moreover, we were able to demonstrate in vivo reduction of endogenous TLR4 expression for at least 1 week, using a similar vector expressing an siRNA for TLR4 in the muscle. In this study, we demonstrate that in vivo suppression of both endogenous (for at least 1 week) and introduced genes (>100 days) is feasible via plasmid-driven siRNA expression after electroporation-mediated intramuscular gene transfer. With this approach the short-term effect of oligonucleotides and the drawbacks of viral gene delivery, like immunological responses, could be circumvented. Therefore, this application of RNA interference is a useful tool with which to investigate gene function and might be promising as a therapeutic tool for locally acting diseases such as restenosis or tumors.


Assuntos
Inativação Gênica , Plasmídeos/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Animais , Bovinos , Linhagem Celular Transformada , Transformação Celular Viral , Eletroporação , Estudos de Viabilidade , Genes Reporter , Humanos , Hipoxantina Fosforribosiltransferase/genética , Lipopolissacarídeos/farmacologia , Luciferases/metabolismo , Medições Luminescentes , Masculino , Camundongos , Camundongos Endogâmicos , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Células NIH 3T3 , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Fatores de Tempo , Receptor 4 Toll-Like/metabolismo
8.
Atherosclerosis ; 193(2): 335-42, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17087966

RESUMO

OBJECTIVE: Inflammatory factors are thought to play a regulatory role in restenosis. Interleukin-10 (IL10) is an important anti-inflammatory cytokine with anti-atherogenic potentials. The aim of this study was to assess the effects of IL10 modulation on cuff-induced neointima formation in hypercholesterolemic APOE*3-Leiden mice. METHODS: The involvement of IL10 in neointima formation was studied in a hypercholesterolemic mouse model of cuff-induced stenosis of the femoral artery by IL10 knocking-out or overexpression procedures. IL10(+/-) mice were crossbred with APOE*3-Leiden mice to generate hypercholesterolemic APOE*3-LeidenIL10(-/-) mice. To achieve IL10 overexpression in APOE*3-Leiden mice, a single intramuscular injection of a murine IL10 overexpression plasmid was performed followed by electroporation. RESULTS: Knocking-out IL10, in hypercholesterolemic APOE*3-Leiden mice, resulted in a significant 1.9-fold increase of neointima surface as compared to APOE*3-LeidenIL10(+/+) littermates (p=0.02). Conversely, a marked 45% inhibition on cuff-induced neointima formation was obtained after IL10 overexpression (p=0.02). Electrodelivery of IL10 vector leads to detectable IL10 serum levels, with a sustained expression over the experimental period of 3 weeks. IL10 overexpression reduced plasma cholesterol levels in APOE*3-Leiden mice, whereas IL10 deficiency in these mice did not lead to altered cholesterol levels as compared to the IL10(+/+) group. Finally, IL10 overexpression stimulated endogenous IL10 mRNA expression in the spleen and reduced the transcriptional responses of several pro-inflammatory cytokines. CONCLUSION: Here, we clearly demonstrate the role of IL10 in the development of neointima formation in hypercholesterolemic mice and the potential therapeutic effect of non-viral electrodelivery of IL10 cDNA to inhibit post-angioplasty restenosis.


Assuntos
Hipercolesterolemia/imunologia , Interleucina-10/imunologia , Túnica Íntima/imunologia , Doenças Vasculares/imunologia , Animais , Modelos Animais de Doenças , Hipercolesterolemia/genética , Interleucina-10/biossíntese , Camundongos , Camundongos Knockout , Doenças Vasculares/genética
9.
J Gen Virol ; 87(Pt 10): 2891-2899, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16963747

RESUMO

Recombinant adenoviral vectors based on type 5 (rAd5) show great promise as a vaccine carrier. However, neutralizing activity against Ad5 is prevalent and high-titred among human populations, and significantly dampens Ad5-based vaccine modalities. The generation of alternative adenoviral vectors with low seroprevalence thus receives much research attention. Here, it is shown that a member from human adenovirus subgroup D, i.e. Ad49, does not cross-react with Ad5 neutralizing activity, making it a candidate serotype for vector development. Therefore, a plasmid system that allows formation of replication-incompetent adenovirus serotype 49 vaccine vectors (rAd49) was constructed and it was demonstrated that rAd49 can be successfully propagated to high titres on existing Ad5.E1-complementing cell lines such as PER.C6. Using an rAd49 vector carrying the luciferase marker gene, detailed seroprevalence studies were performed, demonstrating that rAd49 has low seroprevalence and neutralizing antibody titres worldwide. Also, we have initiated rAd49 vector receptor usage suggesting that rAd49 utilizes hCD46 as a cellular receptor. Finally, the immunogenicity of the rAd49 vector was assessed and it was shown that an rAd49.SIVGag vaccine induces strong anti-SIVGag CD8+ T-lymphocytes in naïve mice, albeit less than an rAd5.SIVGag vaccine. However, in mice with high anti-Ad5 immunity the rAd5.SIVGag vaccine was severely blunted, whereas the anti-SIVGag response was not significantly suppressed using the rAd49.SIVGag vaccine. These data demonstrate the potential of a replication deficient human group D adenoviral vector for vaccination purposes.


Assuntos
Adenovírus Humanos/crescimento & desenvolvimento , Adenovírus Humanos/imunologia , Replicação Viral , Adenovírus Humanos/genética , Adenovírus Humanos/fisiologia , Animais , Anticorpos Antivirais , Linhagem Celular , Engenharia Genética , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Humanos , Proteína Cofatora de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Especificidade de Órgãos , Vacinas Sintéticas , Vacinas Virais/imunologia
10.
Cardiovasc Res ; 68(1): 136-43, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15963479

RESUMO

BACKGROUND: Furin-like proprotein convertases (PCs) are proteolytic activators of proproteins, like membrane type 1-matrix metalloproteinase (MT1-MMP) and transforming growth factor beta (TGF-beta), that are described in the arterial response to injury. However, the involvement of furin-like PCs in the arterial response to injury has not been studied yet. We studied furin, MT1-MMP, MMP levels and TGF-beta signaling after arterial injury. We also investigated the effect of an inhibitor of furin-like PCs, alpha1-antitrypsin Portland (alpha1-PDX), on arterial injury following balloon dilation. METHODS AND RESULTS: NZW rabbit femoral and iliac arteries (N=42) were balloon dilated unilaterally and harvested after 2, 7, 14, 28 or 42 days. Furin mRNA levels were increased after 2 and 7 days. MMP-2 and MT1-MMP levels were increased after day 7 and TGF-beta signaling, by phosphorylating Smad 1/5 and 2/3, was increased at all time points. Inhibition of furin-like PCs, by adenoviral over-expression of alpha1-PDX, blocked proTGF-beta activation and Smad phosphorylation, and reduced MT1-MMP and MMP-2 activation (N=5). In vivo adventitial inhibition of furin-like PCs (N=9) resulted in a reduction of 13.1+/-5.2% in advential and 23.6+/-7.9% in intimal areas (P<0.05), but had no effect on lumen size due to decreased vessel areas. CONCLUSIONS: This study demonstrates that furin-like PCs are involved in the arterial response to injury possibly through activation of the TGF-beta-Smad signaling pathway and identifies furin-like PCs as a possible target to inhibit intimal hyperplasia.


Assuntos
Aterosclerose/metabolismo , Cateterismo , Artéria Femoral/lesões , Furina/fisiologia , Artéria Ilíaca/lesões , Adenoviridae/genética , Animais , Ativação Enzimática , Artéria Femoral/metabolismo , Furina/antagonistas & inibidores , Vetores Genéticos/administração & dosagem , Artéria Ilíaca/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinases da Matriz/metabolismo , Metaloproteinases da Matriz Associadas à Membrana , Modelos Animais , Coelhos , Proteínas Smad Reguladas por Receptor/metabolismo , Transdução Genética/métodos , Fator de Crescimento Transformador beta/metabolismo , alfa 1-Antitripsina/genética
11.
J Clin Endocrinol Metab ; 89(11): 5828-36, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15531549

RESUMO

In the endometrium, angiogenesis is a physiological process, whereas in most adult tissues neovascularization is initiated only during tissue repair or pathological conditions. Pericellular proteolysis plays an important role in angiogenesis being required for endothelial cell migration, invasion, and tube formation. We studied the expression of proteases by human endometrial microvascular endothelial cells (hEMVECs) and their involvement in the formation of capillary tubes and compared these requirements with those of foreskin MVECs (hFMVECs). Inhibition of urokinase and matrix metalloproteinase (MMP) both reduced tube formation in a fibrin or fibrin/collagen matrix. hEMVECs expressed various MMP mRNAs and proteins; in particular MMP-1, MMP-2, and membrane-type (MT)1-, MT3-, and MT4-MMPs. MT3- and MT4-MMP mRNA expressions were significantly higher in hEMVECs than in hFMVECs. Other MT-MMP mRNAs and MMP-9 were hardly detectable. Immunohistochemistry confirmed the presence of MT3-MMP in endothelial cells of endometrial tissue. Overexpression of tissue inhibitor of MMP (TIMP)-1 or TIMP-3 by adenoviral transduction of hEMVECs reduced tube formation to the same extent, whereas only TIMP-3 was able to inhibit tube formation by hFMVECs. Tube formation by hEMVECs was partly inhibited by the presence of anti-MT3-MMP IgG. Thus, in contrast to tube formation by hFMVECs, which largely depends on MT1-MMP, capillary-like tube formation by hEMVECs is, at least in part, regulated by MT3-MMP.


Assuntos
Endométrio/irrigação sanguínea , Células Endoteliais/fisiologia , Metaloendopeptidases/fisiologia , Neovascularização Fisiológica , Adenoviridae/genética , Células Cultivadas , Colágeno Tipo I/farmacologia , Feminino , Fibrinolisina/fisiologia , Humanos , Metaloproteinase 16 da Matriz , Metaloproteinases da Matriz Associadas à Membrana , Inibidor Tecidual de Metaloproteinase-1/genética , Inibidor Tecidual de Metaloproteinase-3/genética , Ativador de Plasminogênio Tipo Uroquinase/fisiologia
12.
J Virol ; 78(23): 13207-15, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15542673

RESUMO

A novel plasmid-based adenovirus vector system that enables manufacturing of replication-incompetent (DeltaE1) adenovirus type 11 (Ad11)-based vectors is described. Ad11 vectors are produced on PER.C6/55K cells yielding high-titer vector batches after purification. Ad11 seroprevalence proves to be significantly lower than that of Ad5, and neutralizing antibody titers against Ad11 are low. Ad11 seroprevalence among human immunodeficiency virus-positive (HIV(+)) individuals is as low as that among HIV(-) individuals, independent of the level of immune suppression. The low level of coinciding seroprevalence between Ad11 and Ad35 in addition to a lack of correlation between high neutralizing antibody titers towards either adenovirus strongly suggest that the limited humoral cross-reactive immunity between these two highly related B viruses appears not to preclude the use of both vectors in the same individual. Ad11 transduces primary cells including smooth muscle cells, synoviocytes, and dendritic cells and cardiovascular tissues with higher efficiency than Ad5. Ad11 and Ad35 appear to have a similar tropism as judged by green fluorescent protein expression levels determined by using a panel of cancer cell lines. In addition, Ad5 preimmunization did not significantly affect Ad11-mediated transduction in C57BL/6 mice. We therefore conclude that the Ad11-based vector represents a novel and useful candidate gene transfer vehicle for vaccination and gene therapy.


Assuntos
Adenovírus Humanos/genética , Terapia Genética , Vetores Genéticos/genética , Replicação Viral , Adenovírus Humanos/imunologia , Adulto , Idoso , Animais , Anticorpos Antivirais/sangue , Antígenos CD/análise , Reações Cruzadas , Vetores Genéticos/imunologia , Humanos , Proteína Cofatora de Membrana , Glicoproteínas de Membrana/análise , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Estudos Soroepidemiológicos , Tropismo , Vacinação
13.
Circ Res ; 95(1): 58-66, 2004 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-15155530

RESUMO

Therapeutic angiogenesis using vascular endothelial growth factor (VEGF) is considered a promising new therapy for patients with arterial obstructive disease. Clinical improvements observed consist of improved muscle function and regression of rest pain or angina. However, direct evidence for improved vascularization, as evaluated by angiography, is weak. In this study, we report an angiogenesis-independent effect of VEGF on ischemic skeletal muscle, ie, upregulation of myoglobin after VEGF treatment. Mice received intramuscular injection with adenoviral VEGF-A or either adenoviral LacZ or PBS as control, followed by surgical induction of acute hindlimb ischemia at day 3. At day 6, capillary density was increased in calf muscle of Ad.VEGF-treated versus control mice (P<0.01). However, angiographic score of collateral arteries was unchanged between Ad.VEGF-treated and control mice. More interestingly, an increase in myoglobin was observed in Ad.VEGF-treated mice. Active myoglobin was 1.5-fold increased in calf muscle of Ad.VEGF-treated mice (P< or =0.01). In addition, the number of myoglobin-stained myofibers was 2.6-fold increased in Ad.VEGF-treated mice (P=0.001). Furthermore, in ischemic muscle of 15 limb amputation patients, VEGF and myoglobin were coexpressed. Finally, in cultured C2C12 myotubes treated with rhVEGF, myoglobin mRNA was 2.8-fold raised as compared with PBS-treated cells (P=0.02). This effect could be blocked with the VEGF receptor tyrosine kinase inhibitor SU5416. In conclusion, we show that VEGF upregulates myoglobin in ischemic muscle both in vitro and in vivo. Increased myoglobin expression in VEGF-treated muscle implies an improved muscle oxygenation, which may, at least partly, explain observed clinical improvements in VEGF-treated patients, in the absence of improved vascularization.


Assuntos
Isquemia/metabolismo , Músculo Esquelético/irrigação sanguínea , Mioglobina/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética , Adenoviridae/genética , Idoso , Amputação Cirúrgica , Animais , Capilares/crescimento & desenvolvimento , Feminino , Expressão Gênica , Terapia Genética , Vetores Genéticos , Humanos , Isquemia/diagnóstico por imagem , Isquemia/terapia , Masculino , Camundongos , Pessoa de Meia-Idade , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/anatomia & histologia , Músculo Esquelético/metabolismo , Mioglobina/genética , Neovascularização Fisiológica , RNA Mensageiro/biossíntese , Radiografia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia
14.
J Virol ; 77(15): 8263-71, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12857895

RESUMO

Replication-deficient human adenovirus type 5 (Ad5) can be produced to high titers in complementing cell lines, such as PER.C6, and is widely used as a vaccine and gene therapy vector. However, preexisting immunity against Ad5 hampers consistency of gene transfer, immunological responses, and vector-mediated toxicities. We report the identification of human Ad35 as a virus with low global prevalence and the generation of an Ad35 vector plasmid system for easy insertion of heterologous genes. In addition, we have identified the minimal sequence of the Ad35-E1B region (molecular weight, 55,000 [55K]), pivotal for complementation of fully E1-lacking Ad35 vector on PER.C6 cells. After stable insertion of the 55K sequence into PER.C6 cells a cell line was obtained (PER.C6/55K) that efficiently transcomplements both Ad5 and Ad35 vectors. We further demonstrate that transduction with Ad35 is not hampered by preexisting Ad5 immunity and that Ad35 efficiently infects dendritic cells, smooth muscle cells, and synoviocytes, in contrast to Ad5.


Assuntos
Adenovírus Humanos/imunologia , Adenovírus Humanos/fisiologia , Vetores Genéticos , Replicação Viral , Proteínas E1B de Adenovirus/química , Proteínas E1B de Adenovirus/genética , Adenovírus Humanos/genética , Animais , Anticorpos Antivirais/imunologia , Linhagem Celular , Células Cultivadas , Células Dendríticas/virologia , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Vetores Genéticos/imunologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Músculo Liso/citologia , Músculo Liso/virologia , Testes de Neutralização , Plasmídeos , Membrana Sinovial/citologia , Membrana Sinovial/virologia , Vacinação , Montagem de Vírus
15.
Arterioscler Thromb Vasc Biol ; 23(9): 1535-40, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12842839

RESUMO

OBJECTIVE: Endothelial cells play a pivotal role in vascular homeostasis. In this study, we investigated the function of the nerve growth factor-induced protein-B (NGFI-B) subfamily of nuclear receptors comprising the TR3 orphan receptor (TR3), mitogen-induced nuclear orphan receptor (MINOR), and nuclear orphan receptor of T cells (NOT) in endothelial cells. METHODS AND RESULTS: The mRNA expression of TR3, MINOR, and NOT in atherosclerotic lesions was assessed in human vascular specimens. Each of these factors is expressed in smooth muscle cells, as described before, and in subsets of endothelial cells, implicating that they might affect endothelial cell function. Adenoviral overexpression of TR3 in cultured endothelial cells resulted in decreased [3H]thymidine incorporation, whereas a dominant-negative TR3 variant that inhibits the activity of endogenous TR3-like factors enhanced DNA synthesis. TR3 interfered with progression of the cell cycle by upregulating p27Kip1 and downregulating cyclin A, whereas expression levels of a number of other cell cycle-associated proteins remained unchanged. CONCLUSIONS: These findings demonstrate that TR3 is a modulator of endothelial cell proliferation and arrests endothelial cells in the G1 phase of the cell cycle by influencing cell cycle protein levels. We hypothesize involvement of TR3 in the maintenance of integrity of the vascular endothelium.


Assuntos
Ciclo Celular/fisiologia , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Receptores de Esteroides/biossíntese , Receptores de Esteroides/fisiologia , Receptores dos Hormônios Tireóideos/biossíntese , Receptores dos Hormônios Tireóideos/fisiologia , Adenoviridae/genética , Divisão Celular/genética , Divisão Celular/fisiologia , Células Cultivadas , DNA Complementar/genética , Endotélio Vascular/virologia , Regulação da Expressão Gênica/genética , Técnicas de Transferência de Genes , Variação Genética/genética , Humanos , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares , Peptídeos/genética , Peptídeos/fisiologia , Estrutura Terciária de Proteína/genética , Receptores de Esteroides/deficiência , Receptores de Esteroides/genética , Receptores dos Hormônios Tireóideos/deficiência , Receptores dos Hormônios Tireóideos/genética , Deleção de Sequência/genética , Células-Tronco/química , Células-Tronco/metabolismo , Células-Tronco/virologia , Ativação Transcricional/genética , Veias Umbilicais/citologia , Veias Umbilicais/virologia , Replicação Viral/genética
16.
Blood ; 101(10): 3933-9, 2003 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-12522008

RESUMO

It has been established that low-density lipoprotein receptor-related protein (LRP) is involved in the cellular uptake and degradation of coagulation factor VIII (FVIII) in vitro. To address the physiologic role of LRP in regulating plasma FVIII in vivo, we used cre/loxP-mediated conditional LRP- deficient mice (MX1cre(+)LRP(flox/flox)). Upon inactivation of the LRP gene, MX1cre(+)LRP(flox/flox) mice had significantly higher plasma FVIII as compared with control LRP(flox/flox) mice (3.4 and 2.0 U/mL, respectively; P <.001). Elevated plasma FVIII levels in MX1cre(+)LRP(flox/flox) mice coincided with increased plasma von Willebrand factor (VWF) (2.0 and 1.6 U/mL for MX1cre(+)LRP(flox/flox) and control LRP(flox/flox) mice, respectively; P <.05). Elevation of plasma FVIII and VWF persisted for at least 6 weeks after inactivation of the LRP gene. Upon comparing plasma FVIII and VWF in individual mice, we observed an increase of the FVIII/VWF ratio in MX1cre(+)LRP(flox/flox) mice as compared with control LRP(flox/flox) mice. Administration of either a vasopressin analog or an endotoxin resulted in increased plasma VWF, but not FVIII. In clearance experiments, MX1cre(+)LRP(flox/flox) mice displayed a 1.5-fold prolongation of FVIII mean residence time. Adenovirus-mediated overexpression of the 39-kDa receptor-associated protein (RAP) in normal mice resulted in a 3.5-fold increase of plasma FVIII. These data confirm that the regulation of plasma FVIII in vivo involves a RAP-sensitive mechanism. Surprisingly, plasma FVIII in MX1cre(+)LRP(flox/flox) mice increased 2-fold after RAP gene transfer. We propose that RAP-sensitive determinants other than hepatic LRP contribute to the regulation of plasma FVIII in vivo.


Assuntos
Fator VIII/metabolismo , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/deficiência , Fator de von Willebrand/metabolismo , Adenoviridae/genética , Animais , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Vetores Genéticos , Genótipo , Humanos , Cinética , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout
17.
Blood ; 101(5): 1810-7, 2003 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-12393408

RESUMO

Adult angiogenesis, associated with pathologic conditions, is often accompanied by the formation of a fibrinous exudate. This temporary matrix consists mainly of fibrin but is intermingled with plasma proteins and collagen fibers. The formation of capillary structures in a fibrinous matrix in vivo was mimicked by an in vitro model, in which human microvascular endothelial cells (hMVECs) seeded on top of a fibrin-10% collagen matrix form capillarylike tubular structures after stimulation with basic fibroblast growth factor/tumor necrosis factor alpha (bFGF/TNF-alpha) or vascular endothelial growth factor (VEGF)/TNF-alpha. In the fibrin-collagen matrix the metalloproteinase inhibitor BB94 inhibited tubule formation by 70% to 80%. Simultaneous inhibition of plasmin and metalloproteinases by aprotinin and BB94 caused a nearly complete inhibition of tubule formation. Adenoviral transduction of tissue inhibitor of metalloproteinases 1 (TIMP-1) and TIMP-3 into endothelial cells revealed that TIMP-3 markedly inhibited angiogenesis, whereas TIMP-1 had only a minor effect. Immunohistochemical analysis showed the presence of matrix metalloproteinase 1 (MMP-1), MMP-2, and membrane-type 1 (MT1)-MMP, whereas MMP-9 was absent. The endothelial production of these MMPs was confirmed by antigen assays and real-time polymerase chain reaction (PCR). MT1-MMP mRNA was markedly increased in endothelial cells under conditions that induced tubular structures. The presence of MMP-1, MMP-2, and MT1-MMP was also demonstrated in vivo in the newly formed vessels of a recanalized arterial mural thrombus. These data suggest that MMPs, in particular MT-MMPs, play a pivotal role in the formation of capillarylike tubular structures in a collagen-containing fibrin matrix in vitro and may be involved in angiogenesis in a fibrinous exudate in vivo.


Assuntos
Metaloproteinase 1 da Matriz/fisiologia , Metaloproteinase 2 da Matriz/fisiologia , Metaloendopeptidases/fisiologia , Neovascularização Patológica/enzimologia , Neovascularização Fisiológica/fisiologia , Fenilalanina/análogos & derivados , Adenoviridae/genética , Aprotinina/farmacologia , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , Colágeno , Meios de Cultura/farmacologia , Vírus Defeituosos/genética , Sinergismo Farmacológico , Fatores de Crescimento Endotelial/farmacologia , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Fibrina , Fator 2 de Crescimento de Fibroblastos/farmacologia , Vetores Genéticos/genética , Cardiopatias/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Linfocinas/farmacologia , Metaloproteinases da Matriz Associadas à Membrana , Neovascularização Patológica/fisiopatologia , Fenilalanina/farmacologia , Inibidores de Proteases/farmacologia , Tiofenos/farmacologia , Trombose/patologia , Inibidor Tecidual de Metaloproteinase-1/genética , Inibidor Tecidual de Metaloproteinase-1/fisiologia , Inibidor Tecidual de Metaloproteinase-3/genética , Inibidor Tecidual de Metaloproteinase-3/fisiologia , Transdução Genética , Fator de Necrose Tumoral alfa/farmacologia , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
18.
Circulation ; 106(12): 1530-5, 2002 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-12234960

RESUMO

BACKGROUND: Smooth muscle cells (SMCs) play a key role in intimal thickening in atherosclerosis and restenosis. The precise signaling pathways by which the proliferation of SMCs is regulated are largely unknown. The TR3 orphan receptor, the mitogen-induced nuclear orphan receptor (MINOR), and the nuclear receptor of T cells (NOT) are a subfamily of transcription factors belonging to the nuclear receptor superfamily and are induced in activated SMCs. In this study, we investigated the role of these transcription factors in SMC proliferation in atherogenesis. METHODS AND RESULTS: Multiple human vascular specimens at distinct stages of atherosclerosis (lesion types II to V by American Heart Association classification) derived from 14 different individuals were studied for expression of these transcription factors. We observed expression of TR3, MINOR, and NOT in neointimal SMCs, whereas no expression was detected in medial SMCs. Adenovirus-mediated expression of a dominant-negative variant of TR3, which suppresses the transcriptional activity of each subfamily member, increases DNA synthesis and decreases p27(Kip1) protein expression in cultured SMCs. We generated transgenic mice that express this dominant-negative variant or full-length TR3 under control of a vascular SMC-specific promoter. Carotid artery ligation of transgenic mice that express the dominant-negative variant of TR3 in arterial SMCs, compared with lesions formed in wild-type mice, results in a 3-fold increase in neointimal formation, whereas neointimal formation is inhibited 5-fold in transgenic mice expressing full-length TR3. CONCLUSIONS: Our results reveal that TR3 and possibly other members of this transcription factor subfamily inhibit vascular lesion formation. These transcription factors could serve as novel targets in the treatment of vascular disease.


Assuntos
Arteriosclerose/etiologia , Proteínas de Ligação a DNA/fisiologia , Músculo Liso Vascular/metabolismo , Receptores de Esteroides , Receptores dos Hormônios Tireóideos , Fatores de Transcrição/fisiologia , Adenoviridae/genética , Animais , Arteriosclerose/genética , Arteriosclerose/metabolismo , Arteriosclerose/patologia , Cardiotônicos/metabolismo , Artérias Carótidas/cirurgia , DNA/biossíntese , Proteínas de Ligação a DNA/genética , Vetores Genéticos , Humanos , Camundongos , Camundongos Transgênicos , Mutação , Proteínas do Tecido Nervoso , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares , RNA Mensageiro/biossíntese , Receptores Citoplasmáticos e Nucleares/biossíntese , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
19.
Circ Res ; 90(10): 1128-34, 2002 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-12039804

RESUMO

Activin A alters the characteristics of human arterial smooth muscle cells (SMCs) toward a contractile, quiescent phenotype. We hypothesize that activin A may prevent SMC-rich neointimal hyperplasia. Here, we study the effect of adenovirus-mediated expression of activin A on neointima formation in vitro and in vivo. Human saphenous vein organ cultures, in which a neointima is formed spontaneously, were infected either with activin A- or lacZ-adenovirus. Activin A-overexpression reduces neointima formation by 78%, whereas no significant reduction was observed after control infection. In addition, the effect of activin A on neointima formation was assessed in vivo in mice with cuffed femoral arteries. In activin A adenovirus-infected mice (IV injection), neointimal hyperplasia is reduced by 77% compared with the SMC-rich neointima in mock-infected or in noninfected mice. Cultured human saphenous vein SMCs and murine aorta SMCs were incubated with activin A and an increased expression of SM22alpha and SM alpha-actin mRNA, and SM alpha-actin protein was demonstrated. Laser-capture microdissection on sections of cuffed murine arteries and subsequent real-time RT-PCR established in vivo induction of SM alpha-actin mRNA in the media of activin A-treated mice. In summary, activin A inhibits neointima formation in vitro and in vivo by preventing SMC dedifferentiation.


Assuntos
Ativinas/genética , Adenoviridae/genética , Arteriopatias Oclusivas/prevenção & controle , Subunidades beta de Inibinas/genética , Músculo Liso Vascular/metabolismo , Ativinas/metabolismo , Ativinas/farmacologia , Animais , Arteriopatias Oclusivas/metabolismo , Arteriopatias Oclusivas/patologia , Diferenciação Celular , Linhagem Celular , Células Cultivadas , Artéria Femoral/citologia , Artéria Femoral/patologia , Vetores Genéticos , Humanos , Hiperplasia , Subunidades beta de Inibinas/metabolismo , Subunidades beta de Inibinas/farmacologia , Cinética , Camundongos , Contração Muscular , Proteínas Musculares/biossíntese , Proteínas Musculares/genética , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/fisiologia , Técnicas de Cultura de Órgãos , Fenótipo , RNA Mensageiro/biossíntese , Veia Safena/anatomia & histologia , Veia Safena/citologia , Túnica Íntima/patologia
20.
Vascul Pharmacol ; 39(6): 293-301, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14567067

RESUMO

Intimal hyperplasia resulting from vascular injury remains a major obstacle in the long-term success of coronary artery bypass grafts. Inhibition of smooth muscle cell (SMC) proliferation using adenoviral gene transfer of cell cycle inhibitors resulted in reduced neointima formation in various animal models. However, little is known about the effect on human SMCs and neointima formation. Here we report the effects of infection with an adenoviral vector encoding a constitutively active form of the retinoblastoma gene (Ad. delta Rb) on proliferation of human saphenous vein SMCs (HSVSMCs) and neointima formation in organ cultures of human saphenous vein. Proliferation of SMCs was inhibited dose-dependently after infection with Ad. delta Rb. A near-total inhibition was found at an Ad. delta Rb concentration of 10(8) pfu/ml. Organ cultures of human saphenous vein segments were used to evaluate the effect of Ad. delta Rb infection on neointima formation and vein graft disease. Segments cultured for 4 weeks develop a neointima that is morphologically highly similar to early initimal lesions found in pathological vein grafts in vivo. Infection of saphenous vein segments with 2 x 10(9) pfu/ml Ad. delta Rb resulted in a 59% reduction of neointimal area when compared to uninfected counterparts, whereas infection with control adenovirus, Ad.LacZ, had no significant effect. The results of this study show that Ad. delta Rb gene transfer might be an efficient approach to prevent neointima formation in human saphenous vein grafts.


Assuntos
Adenoviridae/genética , Ponte de Artéria Coronária , Oclusão de Enxerto Vascular/patologia , Músculo Liso Vascular/fisiologia , Mutação/genética , Neoplasias da Retina/genética , Retinoblastoma/genética , Divisão Celular/fisiologia , Células Cultivadas , Oclusão de Enxerto Vascular/genética , Heterozigoto , Humanos , Hiperplasia/patologia , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Músculo Liso Vascular/citologia , Técnicas de Cultura de Órgãos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Veia Safena/citologia , Veia Safena/crescimento & desenvolvimento , beta-Galactosidase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA