Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
BJU Int ; 91(7): 691-6, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12699487

RESUMO

OBJECTIVE: To determine whether over-expression of nitric oxide synthase (NOS) in the corpus cavernosum of the penis improves erectile function, as NO is an important transmitter for genitourinary tract function, mediating smooth muscle relaxation and being essential for penile erection. MATERIALS AND METHODS: The inducible form of the enzyme NOS (iNOS) was introduced into the corpus cavernosum of adult Sprague-Dawley rats (250-300 g) by injecting a solution of plasmid, adenovirus or adenovirus-transduced myoblast cells (adeno-myoblasts). Plasmid, adenovirus and adeno-myoblasts encoding the expression of the beta-galactosidase reporter gene were also injected into rats. RESULTS: Throughout the corpora cavernosum there was expression of beta-galactosidase after injecting each of the three solutions. Maximum staining was greatest for adeno-myoblast, then adenovirus and then plasmid. The mean (sd) basal intracavernosal pressure (ICP) of iNOS-treated animals (adenovirus and adeno-myoblast) increased to 55 (23) cmH2O, compared with naive animals with a basal ICP of 5 (6) cmH2O (P = 0.001). Stimulating the cavernosal nerve (15 Hz, 1.5 ms, 10-40 V, 1 min) resulted in a doubling of the ICP (adenovirus and adeno-myoblast) from the basal level of the iNOS-treated animals. Direct in situ measurement of NO showed the release of 1-1.3 micro mol/L in the adeno-myoblast penis. CONCLUSION: Myoblast-mediated gene therapy was more successful for delivering iNOS into the corpus cavernosum than direct adenovirus injection or plasmid transfection. Surprisingly, implanting muscle cells into the penis is not only feasible but also beneficial. Gene therapy for NOS may open new avenues of treatment for erectile dysfunction. Control of NOS expression would be necessary to prevent priapism.


Assuntos
Disfunção Erétil/terapia , Óxido Nítrico Sintase/administração & dosagem , Adenoviridae , Animais , Técnicas de Transferência de Genes , Masculino , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Pênis/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
2.
J Vasc Surg ; 34(5): 878-84, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11700490

RESUMO

OBJECTIVE: Adequate proximal neck length is important for proper endovascular treatment of abdominal aortic aneurysms (AAAs). Placement of endografts in AAAs with relatively short proximal necks may require covering the origin of accessory renal arteries. Exclusion of these arteries carries the theoretical concern of regional renal ischemia associated with loss of parenchyma or worsening hypertension. We reviewed our experience with accessory renal exclusions during endovascular AAA repair to determine the frequency and severity of complications. METHODS: Complete records were available for review on 311 of 325 consecutive patients treated with endovascular grafts for AAAs from February 6, 1996, to March 15, 2001. The presence of accessory renal arteries was ascertained from preoperative/intraoperative aortography or from computed tomographic scanning. Sizes of the accessories were measured by using the main renal arteries as a reference. Considerations for excluding the accessory renal arteries were based on the likelihood of successful proximal attachment to healthy aorta, an accessory vessel whose size does not exceed the diameter of the main renal artery, and the absence of renal disease. RESULTS: The mean follow-up was 11.5 months. Fifty-two accessory renal arteries were documented in 37 patients (12%), ranging from 1 to > or =3 per patient. Of these, 26 accessory renal arteries were covered in 24 patients. Patients ranged in age from 57 to 85 years (mean, 74.1 years), with 20 men and 4 women. The Ancure device was used in 23 patients and the Excluder device in one. Of the accessories excluded, 22 originated above the aneurysm and 4 originated directly from the aneurysm itself. There were no perioperative mortalities. One patient died 5 months after surgery from an unrelated condition. There was one type I (distal) endoleak and no type II endoleaks. Five patients (21%) had segmental renal infarction associated with the side of accessory renal artery exclusion. Only one patient with segmental infarction had significant postoperative hypertension that resulted in changes in blood pressure medication. The blood pressure reverted to normal 3 months later. One patient with a stenotic left main renal artery required exclusion of the accessory renal artery for successful proximal attachment. Serum creatinine levels remained unchanged throughout follow-up in all but one patient, in whom progressive postoperative renal failure developed despite normal renal flow scan, presumably from intraoperative manipulation and contrast nephropathy. CONCLUSION: Exclusion of accessory renal arteries to facilitate endovascular AAA repair appears to be well tolerated. Long-term sequelae seem infrequent and mild.


Assuntos
Aneurisma da Aorta Abdominal/cirurgia , Artéria Renal/anormalidades , Idoso , Implante de Prótese Vascular , Feminino , Seguimentos , Humanos , Masculino , Radiografia , Artéria Renal/diagnóstico por imagem , Fatores de Tempo
3.
Mol Urol ; 5(1): 37-43, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11689150

RESUMO

BACKGROUND AND PURPOSE: Nitric oxide (NO) has been recognized as an important transmitter for genitourinary tract function. This transmitter mediates smooth muscle relaxation and is essential for erection. The objective of our research was to determine whether overexpression of nitric oxide synthase (NOS) in the corpus cavernosum of the penis would correct erectile dysfunction. MATERIALS AND METHODS: We introduced the inducible form of the enzyme NOS (iNOS) into the corpus cavernosum of adult (250-300 g) male Sprague-Dawley rats by injecting a solution of plasmid, adenovirus, or adenovirus-transduced myoblast cells (adeno-myoblast) (N = 3-5 each group). We also injected plasmid, adenovirus, and adeno-myoblast encoding the expression of the beta-gatactosidase reporter gene. RESULTS: We noted expression of beta-galactosidase throughout the corpora cavernosum after injection of each of the three solutions. Staining was greatest for adeno-myoblast followed by adenovirus and then plasmid. The basal intracavernous pressure (ICP) of iNOS-treated animals (adenovirus and adenovirus-transduced myoblast) increased to 55 +/- 23 cm H(2)O v 5 +/- 6 H(2)O in naive animals (P = 0.001). Stimulation of the cavernous nerve (15 Hz, 1.5 msec, 10-40 V, 1 min) resulted in a twofold increase in ICP (adenovirus and adeno-myoblast) from the basal level of the iNOS-treated animals. Direct in situ measurement of NO demonstrated release of 1 to 1.3 microM NO in the adeno-myoblast-treated penis. CONCLUSION: Myoblast-mediated gene therapy was more successful in delivering iNOS into the corpus cavernosum than were the direct adenovirus or plasmid transfection methods. Gene therapy of NOS may open new avenues of treatment for erectile dysfunction. Control of NOS expression would be necessary to prevent priapism.


Assuntos
Disfunção Erétil/terapia , Terapia Genética/métodos , Óxido Nítrico Sintase/genética , Adenoviridae , Animais , Linhagem Celular , Escherichia coli , Vetores Genéticos , Masculino , Óxido Nítrico/análise , Óxido Nítrico Sintase Tipo II , Pênis/enzimologia , Plasmídeos , Ratos , Ratos Sprague-Dawley , Transfecção , beta-Galactosidase/análise , beta-Galactosidase/genética
4.
J Vasc Surg ; 34(1): 156-65, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11436090

RESUMO

OBJECTIVE: The aim of this study is to determine whether adenoviral inducible nitric oxide synthase (iNOS) gene transfer could inhibit intimal hyperplasia (IH) in porcine internal jugular veins interposed into the carotid artery circulation. METHODS: Porcine internal jugular veins were transduced passively with 1 x 10(11) particles of an adenoviral vector carrying either the human iNOS (AdiNOS) or beta-galactosidase (AdlacZ) cDNA for 30 minutes and then interposed into the carotid artery circulation. Segments of each vein graft were maintained in an ex vivo organ culture to measure nitrite accumulation, a marker of nitric oxide synthesis. The grafts were analyzed immunohistochemically for the presence of neutrophils, macrophages, and leukocytes by staining for myeloperoxidase, ED1, and CD45, respectively, at 3 (n = 4) and 7 (n = 4) days. Morphometric analyses and cellular proliferation (Ki67 staining) were assessed at 3 (n = 4), 7 (n = 4), and 21 days (n = 8). RESULTS: AdlacZ-treated vein grafts demonstrated high levels of beta-galactosidase expression at 3 days with a gradual decline thereafter. Nitrite production from AdiNOS-treated vein grafts was approximately fivefold greater than AdlacZ-treated grafts (P =.00001). AdiNOS or AdlacZ treatment was associated with minimal graft inflammation. Cellular proliferation rates were significantly reduced in AdiNOS-treated grafts as compared with controls at both 3 (41%, P =.000004) and 7 days (32%, P =.0001) after bypass. This early antiproliferative effect was most pronounced at the distal anastomosis (65%, P =.0005). The iNOS gene transfer reduced the intimal/medial area ratio in vein grafts at 7 (36%, P =.009) and 21 days (30%, P =.007) versus controls. This inhibition of IH was again more prominent in the distal segments of the grafts (P =.01). CONCLUSION: Adenovirus-mediated iNOS gene transfer to porcine internal jugular vein grafts effectively reduced cellular proliferation and IH. Although iNOS gene transfer reduced IH throughout the entire vein graft, the most pronounced effect was measured at the distal anastomosis. These results suggest potential for iNOS-based genetic modification of vein grafts to prolong graft patency.


Assuntos
Técnicas de Transferência de Genes , Veias Jugulares/transplante , Túnica Íntima/patologia , Adenoviridae/genética , Animais , Hiperplasia , Imuno-Histoquímica , Técnicas In Vitro , Óxido Nítrico Sintase , Óxido Nítrico Sintase Tipo II , Suínos
5.
J Vasc Surg ; 31(6): 1214-28, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10842159

RESUMO

OBJECTIVE: Overexpression of the inducible nitric oxide synthase (iNOS) gene inhibits neointimal hyperplasia after arterial injury. The purpose of this study was to examine the mechanism by which nitric oxide (NO) inhibits vascular smooth muscle cell (VSMC) proliferation, specifically focusing on signaling pathways known to be activated by NO, including cyclic guanosine monophosphate (cGMP), p53, and p42/44 mitogen-activated protein kinase (MAPK). METHODS AND RESULTS: VSMCs that were subjected to iNOS gene transfer demonstrated a reduction in proliferation (80%) that was associated with a marked increase in p21 expression. The antiproliferative and p21 stimulatory effects of NO were not suppressed by the soluble guanylate cyclase inhibitor ODQ, implicating cGMP-independent signaling. The role of p53 in NO-mediated upregulation of p21 and inhibition of proliferation was evaluated using p53 -/- VSMCs. A similar reduction in cellular proliferation and upregulation of p21 expression were achieved with iNOS gene transfer as well as treatment with the NO-donor S-nitroso-N-acetylpenicillamine (SNAP), demonstrating the p53-independent nature of these NO-mediated pathways. The transfer of the iNOS gene activated the p42/44 MAPK, and inhibition of this MAPK pathway with PD98059 partially blocked the antiproliferative effects of NO and completely inhibited the p21 stimulatory effects of NO. For confirmation that iNOS overexpression upregulated p21 in vivo, injured rat carotid arteries were infected with an adenoviral vector carrying the iNOS gene and demonstrated a marked upregulation of p21 expression at three days. However, the ability of NO to inhibit VSMC proliferation does not solely depend on p21 upregulation since the NO-donor SNAP-inhibited VSMC proliferation in p21 -/- VSMCs. CONCLUSION: Nitric oxide inhibits VSMC proliferation in association with the upregulation of p21; both occur independent of p53 and cGMP while being partially mediated through the p42/44 MAPK signaling cascade. This represents one potential mechanism by which NO inhibits VSMC proliferation.


Assuntos
GMP Cíclico/metabolismo , Regulação Enzimológica da Expressão Gênica , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Músculo Liso Vascular/citologia , Óxido Nítrico Sintase/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima/genética , Animais , Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Divisão Celular/genética , GMP Cíclico/genética , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Técnicas de Transferência de Genes , Guanilato Ciclase/antagonistas & inibidores , Humanos , Hiperplasia , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/genética , Doadores de Óxido Nítrico/farmacologia , Penicilamina/análogos & derivados , Penicilamina/farmacologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/genética , Proteína Supressora de Tumor p53/genética , Túnica Íntima/metabolismo , Túnica Íntima/patologia
7.
Transplantation ; 69(6): 1186-92, 2000 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-10762225

RESUMO

BACKGROUND: Inducible nitric oxide synthase (iNOS) is up-regulated in rejecting allografts and is protective against allograft arteriosclerosis; it suppresses neointimal smooth muscle cell accumulation and inhibits adhesion of platelets and leukocytes to the endothelium. However, the functional importance of endothelial NOS (eNOS) in the rejecting allografts remains unclear. METHODS: We examined the effects of selective eNOS deficiency in aortic allografts in a murine chronic rejection model using grafts from eNOS knockout (KO) mice (C57BL/6 background; H2b) and normal C3H (H2K) as recipients. Grafts from wild-type C57BL/6 mice served as controls. Grafts from iNOS KO mice served as a second group of controls where the contribution from iNOS was eliminated but eNOS was preserved. Aortic grafts were harvested and analyzed at days 10-14, 18-22, and 26-30 after transplantation. RESULTS: Endothelial NOS-deficient grafts showed significantly increased intima/media ratios at days 26-30 compared to controls. Immunostaining demonstrated that in eNOS KO grafts, eNOS was not detectable whereas iNOS was expressed prominently in infiltrating recipient mononuclear cells. In control grafts, eNOS expression was preserved in the endothelium even by day 30, and associated with a decrease in intimal thickening. We further demonstrated that early overexpression of iNOS by ex vivo gene transfer completely prevented the development of arteriosclerosis associated with eNOS deficiency. CONCLUSIONS: We found that eNOS plays a protective role in allografts, and that in eNOS-deficient allografts, early overexpression of iNOS is capable of preventing the development of allograft arteriosclerosis. In allografts with dysfunctional vascular endothelium and impaired eNOS activity as a result of ischemia or native arteriosclerotic disease, iNOS gene therapy may serve to improve their long-term survival and function.


Assuntos
Aorta Torácica/transplante , Arteriosclerose/prevenção & controle , Óxido Nítrico Sintase/uso terapêutico , Animais , Aorta Torácica/metabolismo , Arteriosclerose/etiologia , Rejeição de Enxerto/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase/biossíntese , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , Transplante Homólogo/efeitos adversos , Transplante Homólogo/imunologia
8.
Semin Perinatol ; 24(1): 51-4, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10709860

RESUMO

Nitric oxide (NO) is intimately involved in vascular homeostasis through its antiplatelet, antiproliferative, and vasodilating actions. Because of these beneficial properties, methods of harnessing NO for the prevention of vascular injury responses, such as intimal hyperplasia, are being explored. One such method involves gene transfer of an NO synthase (NOS) to sites of vascular injury to provide for local NO synthesis. Gene delivery of the inducible NOS (iNOS) cDNA to sites of vascular injury in animal models dramatically reduced smooth muscle proliferation and intimal hyperplasia. The cellular mechanisms by which NO inhibits smooth muscle cell proliferation appear to be independent of cyclic guanosine monophosphate production but are linked to the upregulation of the cell cycle inhibitor p21. p21 upregulation occurs independent of p53 expression. Instead, p42/44 mitogen activated protein kinase activation by NO results in reduced cellular proliferation and increased p21 expression, suggesting NO inhibits intimal hyperplasia through cell cycle arrest as mediated by p21 and the signaling pathway involved in p21 upregulation may be regulated by p42/44 mitogen activated protein kinase.


Assuntos
Terapia Genética/métodos , Óxido Nítrico Sintase/genética , Doenças Vasculares/genética , Doenças Vasculares/terapia , Animais , Terapia Genética/tendências , Humanos , Doenças Vasculares/enzimologia
9.
Arch Surg ; 135(2): 191-7, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10668880

RESUMO

BACKGROUND: Adenovirus is widely used as a vector for gene transfer to the vasculature. However, the efficiency of these vectors can be limited by ineffective viral-target cell interactions. Viral attachment, which largely determines adenoviral tropism, is mediated through binding of the adenoviral fiber coat protein to the Coxsackievirus and adenovirus receptor, while internalization follows binding of the adenoviral RGD motif to alpha(v)-integrin receptors. Modifications of the fiber coat protein sequence have been successful for targeting the adenovirus to more prevalent receptors in the vasculature, including heparan sulfate-containing receptors and alpha(v)-integrin receptors. HYPOTHESIS: Modified adenoviral vectors targeted to receptors more prevalent in the vasculature result in an increased transfer efficiency of the virus in vitro and in vivo even in the presence of clinically relevant doses of heparin. DESIGN: We tested 2 modified E1- and E3-deleted Ad5 type adenoviral vectors containing the beta-galactosidase gene. AdZ.F(pK7) contains multiple positively charged lysines in the fiber coat protein that target the adenovirus to heparan sulfate receptors, while AdZ.F(RGD) contains an RGD integrin-binding sequence in the fiber coat protein that allows binding to alpha(v)-integrin receptors. The gene transfer efficiency of these modified viruses was compared in rat aortic smooth muscle cells in vitro and in an in vivo porcine model of balloon-induced arterial injury. Because of the use of heparin during most vascular surgical procedures and the concern that heparin might interfere with the binding of AdZ.F(pK7) to heparan sulfate receptors, the effect of heparin on the in vitro and in vivo transfer efficiency of these 2 modified adenoviruses was evaluated. RESULTS: In vitro infection of rat aortic smooth muscle cells with AdZ.F(pK7) and AdZ.F(RGD) resulted in significantly higher levels of beta-galactosidase expression compared with the unmodified adenovirus (mean +/- SEM, 1766.3 +/- 89.1 and 44.8 +/- 3.4 vs 10.1 +/- 0.7 mU per milligram of protein; P<.001). Following heparin administration, the gene transfer efficiency achieved with AdZ.F(pK7) diminished slightly in a concentration-dependent manner. However, the transfer efficiency was still greater than with the unmodified virus (mean +/- SEM, 1342.3 +/- 101.8 vs 4.8 +/- 0.4 mU per milligram of protein; P<.001). In vivo, following injury to the pig iliac artery with a 4F Fogarty balloon catheter, we found that AdZ.F(pK7) transduced the artery approximately 35-fold more efficiently than AdZ.F and 3-fold more efficiently than AdZ.F(RGD) following the administration of intravenous heparin, 100 U/kg body weight, and heparinized saline irrigation. CONCLUSIONS: Modifications of the adenovirus that lead to receptor targeting resulted in significantly improved gene transfer efficiencies. These improvements in transfer efficiencies observed with the modified vectors decreased slightly in the presence of heparin. However, AdZ.F(pK7) was still superior to AdZ.F(RGD) and AdZ.F despite heparin administration. These data demonstrate that modifications of adenoviral vectors that enhance binding to heparan sulfate receptors significantly improve gene transfer efficiency even in the presence of heparin and suggest an approach to optimize gene transfer into blood vessels.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética/métodos , Adenoviridae , Animais , Vetores Genéticos , Heparina/farmacologia , Técnicas In Vitro , Masculino , Músculo Liso Vascular , Ratos , Ratos Sprague-Dawley , Suínos , beta-Galactosidase/genética
10.
J Vasc Surg ; 31(2): 364-74, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10664504

RESUMO

PURPOSE: We have shown that gene transfer of the inducible nitric oxide synthase (iNOS) gene to injured arteries inhibits the development of intimal hyperplasia. One mechanism by which nitric oxide (NO) may inhibit this process is through the upregulation of the cyclin-dependent kinase inhibitor p21, which induces a G0/G1 cell cycle arrest, leading to an inhibition of vascular smooth muscle cell (VSMC) proliferation. Because NO induced such a dramatic upregulation of p21 and because p21 is a universal inhibitor of the cell cycle, this study aimed to determine how NO upregulates p21 protein expression in VSMCs. METHODS: p21 messenger RNA (mRNA) levels in rat aortic smooth muscle cells (RASMCs) were determined by Northern blot analysis after treatment with S-nitroso-N-acetylpenicillamine (SNAP) or after adenoviral iNOS gene transfer. p21 protein levels in RASMCs in similar conditions were determined by Western blot analysis. Levels of ubiquinated p21 in these same treatment groups were assessed by immunoprecipitation of p21 from RASMCs, followed by western blot analysis for ubiquitin. Protein tyrosine and protein serine/threonine phosphatase activity after treatment with SNAP, plus or minus the phosphatase inhibitors calyculin A or cantharidin, were measured with (32)P-labeled myelin basic protein as a substrate. RESULTS: NO exposure by the NO-donor SNAP or iNOS gene transfer induced a dose- and time-dependent increase in p21 protein expression in RASMCs. p21 mRNA levels were significantly increased after SNAP treatment only at the 6-hour point, but were not increased at 24 hours. In contrast, protein levels were increased from 6 to 24 hours, and transcriptional inhibitors did not inhibit this increase in protein synthesis. The increase in p21 protein expression induced by NO was associated with less of the ubiquinated form of p21 at both early and late points. Furthermore, NO induced an increase in both protein tyrosine and protein serine/threonine phosphatase activity. Inhibition of these phosphatases with calyculin A or cantharidin prevented the upregulation of p21 protein expression by NO. CONCLUSION: These data indicate that one mechanism by which NO upregulates p21 protein expression is through the prevention of p21 protein degradation by the ubiquitin-proteasome pathway in association with increased protein tyrosine and serine/threonine phosphatase activity.


Assuntos
Adenosina Trifosfatases/efeitos dos fármacos , Ciclinas/efeitos dos fármacos , Cisteína Endopeptidases/efeitos dos fármacos , Inibidores Enzimáticos , Complexos Multienzimáticos/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Óxido Nítrico/farmacologia , Ubiquitinas/efeitos dos fármacos , Vasodilatadores/farmacologia , Adenosina Trifosfatases/metabolismo , Animais , Aorta Torácica/citologia , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/metabolismo , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Cisteína Endopeptidases/metabolismo , Inibidores Enzimáticos/metabolismo , Complexos Multienzimáticos/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Complexo de Endopeptidases do Proteassoma , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Transdução Genética , Ubiquitinas/metabolismo , Regulação para Cima/efeitos dos fármacos
11.
Surgery ; 126(2): 323-9, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10455901

RESUMO

BACKGROUND: Vein graft failure as the result of intimal hyperplasia (IH) remains a significant clinical problem. Ex vivo modification of vein grafts using gene therapy is an attractive approach to attenuate IH. Gene transfer of the inducible nitric oxide synthase (iNOS) gene effectively reduces IH. However, iNOS activity after gene transfer may be impaired by the availability of cofactor, such as tetrahydrobiopterin (BH4). The purpose of this study is to determine the optimal conditions for ex vivo adenoviral-mediated iNOS gene transfer into arterial and venous vessels. METHODS: Porcine internal jugular veins and carotid arteries were infected ex vivo with the adenoviral iNOS vector (AdiNOS) and with an adenovirus carrying the cDNA encoding guanosine triphosphate cyclohydrolase I (AdGTPCH), the rate-limiting enzyme for BH4 synthesis. The production of nitrite, cyclic guanosine monophosphate (cGMP), and biopterin were assessed daily. RESULTS: Nitric oxide (NO) production after iNOS gene transfer was maximal when vessels were cotransduced with AdGTPCH. NO production in these vessels persisted for more than 10 days. Vein segments generated approximately 2-fold more nitrite, cGMP, and biopterin than arterial segments infected with AdiNOS/AdGTPCH. Submerging vein segments into adenoviral solution resulted in improved gene transfer with greater nitrite and cGMP release compared with infections carried out under pressure intraluminally. Similarly, injury to the vein segments before infection with AdiNOS resulted in less nitrite production. CONCLUSIONS: These data demonstrate that AdiNOS can efficiently transduce vein segments ex vivo and that the cotransfer of GTPCH can optimize iNOS enzymatic activity. This cotransfer technique may be used to engineer vein grafts before coronary artery bypass to prevent IH.


Assuntos
Técnicas de Transferência de Genes , Óxido Nítrico Sintase/genética , Veias/metabolismo , Veias/transplante , Adenoviridae/genética , Animais , Biopterinas/biossíntese , GMP Cíclico/biossíntese , GTP Cicloidrolase/genética , Masculino , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo II , Técnicas de Cultura de Órgãos , Suínos
13.
Curr Opin Nephrol Hypertens ; 8(1): 75-81, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9914863

RESUMO

A wide array of vascular disorders have been shown to benefit from gene therapy. Given the vasoprotective role that nitric oxide plays in the vasculature, it is understandable why gene therapy with the three different isoforms of nitric oxide synthase has been so successful. This review summarizes the current literature pertaining to nitric oxide synthase gene transfer to the vascular wall.


Assuntos
Vasos Sanguíneos/fisiologia , Técnicas de Transferência de Genes , Óxido Nítrico Sintase/genética , Animais , Humanos
14.
J Leukoc Biol ; 65(1): 50-8, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9886246

RESUMO

Nitric oxide (NO) produced by inducible NO synthase (iNOS, NOS-2) is an important component of the macrophage-mediated immune defense toward numerous pathogens. Murine macrophages produce NO after cytokine activation, whereas, under similar conditions, human macrophages produce low levels or no NO at all. Although human macrophages can express iNOS mRNA and protein on activation, whether they possess the complete machinery necessary for NO synthesis remains controversial. To define the conditions necessary for human monocytes/macrophages to synthesize NO when expressing a functional iNOS, the human monocytic U937 cell line was engineered to synthesize this enzyme, following infection with a retroviral expression vector containing human hepatic iNOS (DFGiNOS). Northern blot and Western blot analysis confirmed the expression of iNOS in transfected U937 cells both at the RNA and protein levels. NOS enzymatic activity was demonstrated in cell lysates by the conversion of L-[3H]arginine into L-[3H]citrulline and the production of NO by intact cells was measured by nitrite and nitrate accumulation in culture supernatants. When expressing functional iNOS, U937 cells were capable of releasing high levels of NO. NO production was strictly dependent on supplementation of the culture medium with tetrahydrobiopterin (BH4) and was not modified by stimulation of the cells with different cytokines. These observations suggest that (1) human monocytic U937 cells contain all the cofactors necessary for NO synthesis, except BH4 and (2) the failure to detect NO in cytokine-stimulated untransfected U937 cells is not due to the presence of a NO-scavenging molecule within these cells nor to the destabilization of iNOS protein. DFGiNOS U937 cells represent a valuable human model to study the role of NO in immunity toward tumors and pathogens.


Assuntos
Monócitos/enzimologia , Óxido Nítrico Sintase/biossíntese , Óxido Nítrico/biossíntese , Animais , Boroidretos/farmacologia , Linhagem Celular , DNA Complementar/genética , DNA Complementar/metabolismo , Humanos , Fígado/enzimologia , Macrófagos/enzimologia , Camundongos , Monócitos/metabolismo , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Células U937
15.
Cardiovasc Res ; 43(3): 650-7, 1999 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-10690336

RESUMO

The role nitric oxide (NO) plays in the cardiovascular system is complex and diverse. Even more controversial is the role that the inducible NO synthase enzyme (iNOS) serves in mediating different aspects of cardiovascular pathophysiology. Following arterial injury, NO has been shown to serve many vasoprotective roles, including inhibition of platelet aggregation and adherence to the site of injury, inhibition of leukocyte adherence, inhibition of vascular smooth muscle cell (VSMC) proliferation and migration, and stimulation of endothelial cell (EC) growth. These properties function together to preserve a normal vascular environment following injury. In this review, we discuss what is known about the involvement of iNOS in the vascular injury response. Additionally, we discuss the beneficial role of iNOS gene transfer to the vasculature in preventing the development of neointimal thickening. Lastly, the pathophysiology of transplant vasculopathy is discussed as well as the role of iNOS in this setting.


Assuntos
Cateterismo/efeitos adversos , Endotélio Vascular/lesões , Músculo Liso Vascular/lesões , Óxido Nítrico Sintase/fisiologia , Óxido Nítrico/metabolismo , Animais , Artérias , Divisão Celular , Movimento Celular , Endotélio Vascular/metabolismo , Indução Enzimática , Terapia Genética , Transplante de Coração , Humanos , Músculo Liso Vascular/metabolismo , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Agregação Plaquetária
16.
Nat Med ; 4(12): 1371-6, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9846573

RESUMO

The finding of frequent nitric oxide synthase expression in human cancers indicates that nitric oxide has a pathophysiological role in carcinogenesis. To determine the role of nitric oxide in tumor progression, we generated human carcinoma cell lines that produced nitric oxide constitutively. Cancer cells expressing inducible nitric oxide synthase that had wild-type p53 had reduced tumor growth in athymic nude mice, whereas those with mutated p53 had accelerated tumor growth associated with increased vascular endothelial growth factor expression and neovascularization. Our data indicate that tumor-associated nitric oxide production may promote cancer progression by providing a selective growth advantage to tumor cells with mutant p53, and that inhibitors of inducible nitric oxide synthase may have therapeutic activity in these tumors.


Assuntos
Fatores de Crescimento Endotelial/fisiologia , Linfocinas/fisiologia , Neoplasias Experimentais/enzimologia , Neoplasias Experimentais/patologia , Óxido Nítrico Sintase/biossíntese , Proteína Supressora de Tumor p53/fisiologia , Animais , Apoptose , Técnicas de Transferência de Genes , Humanos , Camundongos , Transplante de Neoplasias , Neovascularização Patológica , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Molécula-1 de Adesão Celular Endotelial a Plaquetas/análise , Transplante Heterólogo , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
17.
Am J Physiol ; 275(4): L717-28, 1998 10.
Artigo em Inglês | MEDLINE | ID: mdl-9755104

RESUMO

Our group recently reported that cultured sheep pulmonary artery endothelial cells (SPAECs) became resistant to lipopolysaccharide (LPS)-induced apoptosis several days after constitutive synthesis of nitric oxide (NO) after adenoviral (Ad) transfer of inducible NO synthase (iNOS) or exposure to the NO donor S-nitroso-N-acetylpenicillamine (SNAP) (E. Tzeng, Y.-M. Kim, B. R. Pitt, A. Lizonova, I. Kovesdi, and T. R. Billiar. Surgery 122: 255-263, 1997). In the present study, we confirmed this observation by establishing stable transfectants after retroviral gene transfer [replication-deficient retrovirus (DFG)] of human iNOS (DFG-iNOS) SPAECs and then used all three approaches (Ad, DFG, and SNAP) to determine underlying mechanisms of this phenomenon. Continuous endogenous production of NO in itself did not cause apoptosis as assessed by phase-contrast microscopy, nuclear morphology, and internucleosomal DNA fragmentation. Prolonged (72-96 h) synthesis of NO, however, after DFG- or replication-deficient adenovirus (Ad. CMV)-iNOS or SNAP (100 microM, 96 h) inhibited LPS-induced apoptosis. The kinetics of such protection suggested that NO may be inducing other gene products. Ad-mediated transfer of manganese superoxide dismutase (MnSOD) decreased the sensitivity of wild-type SPAECs to LPS-induced apoptosis. MnSOD, however, was not induced in an NG-monomethyl-L-arginine (L-NMMA)-sensitive time-dependent fashion after Ad.CMV-iNOS. Other inducible genes that may be affected by NO and that may protect against potential oxidant-mediated LPS-induced apoptosis including 70-kDa heat shock protein, heme oxygenase-1, metallothionein, and Bcl-2 also were not elevated in an L-NMMA-sensitive, time-dependent fashion. Although the candidate gene product underlying NO-induced protection remains unclear, we did note that prolonged synthesis of NO inhibited LPS-induced activation of an interleukin-1beta-converting enzyme-like cysteine protease (cysteine protease protein-32-like) in a dithiothreitol-sensitive fashion, suggesting that S-nitrosylation of an important downstream target of convergence of apoptotic signals may contribute to the sensitivity of SPAECs to LPS.


Assuntos
Apoptose/fisiologia , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Lipopolissacarídeos/farmacologia , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico/fisiologia , Adenoviridae , Animais , Apoptose/efeitos dos fármacos , Caspase 3 , Caspases/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/fisiologia , Núcleo Celular/ultraestrutura , Células Cultivadas , Endotélio Vascular/efeitos dos fármacos , Vetores Genéticos , Humanos , Lipopolissacarídeos/antagonistas & inibidores , Fígado/enzimologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Mitocôndrias/ultraestrutura , Vírus da Leucemia Murina de Moloney , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Nitritos/metabolismo , Artéria Pulmonar , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/metabolismo , Ovinos , Superóxido Dismutase/metabolismo , Transfecção , Vacúolos/ultraestrutura , ômega-N-Metilarginina/farmacologia
18.
J Am Coll Surg ; 187(3): 295-306, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9740187

RESUMO

BACKGROUND: Inadequate nitric oxide (NO) availability may underlie vascular smooth muscle overgrowth that contributes to vascular occlusive diseases including atherosclerosis and restenosis. NO possesses a number of properties that should inhibit this hyperplastic healing response, such as promoting reendothelialization, preventing platelet and leukocyte adherence, and inhibiting cellular proliferation. STUDY DESIGN: We proposed that shortterm but sustained increases in NO synthesis achieved with inducible NO synthase (iNOS) gene transfer at sites of vascular injury would prevent intimal hyperplasia. We constructed an adenoviral vector, AdiNOS, carrying the human iNOS cDNA and used it to express iNOS at sites of arterial injury in vivo. RESULTS: AdiNOS-treated cultured vascular smooth muscle cells produced up to 100-fold more NO than control cells. In vivo iNOS gene transfer, using low concentrations of AdiNOS (2 x 10(6) plaque forming units [PFU]/rat) to injured rat carotid arteries, resulted in a near complete (>95%) reduction in neointima formation even when followed longterm out to 6 weeks post-injury. This protective effect was reversed by the continuous administration of an iNOS selective inhibitor L-N6-(1-iminoethyl)-lysine. However, iNOS gene transfer did not lead to regression of preestablished neointimal lesions. In an animal model more relevant to human vascular healing, iNOS gene transfer (5 x 10(8) PFU/pig) to injured porcine iliac arteries in vivo was also efficacious, reducing intimal hyperplasia by 51.8%. CONCLUSIONS: These results indicate that shortterm overexpression of the iNOS gene initiated at the time of vascular injury is an effective method of locally increasing NO levels to prevent intimal hyperplasia.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética/métodos , Hiperplasia/prevenção & controle , Músculo Liso Vascular/patologia , Óxido Nítrico Sintase/genética , Adenoviridae , Animais , Artérias Carótidas/enzimologia , Artérias Carótidas/patologia , Células Cultivadas , Vetores Genéticos , Artéria Ilíaca/enzimologia , Artéria Ilíaca/patologia , Masculino , Músculo Liso Vascular/enzimologia , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II , Ratos , Ratos Sprague-Dawley , Suínos
19.
Surgery ; 124(2): 278-83, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9706149

RESUMO

BACKGROUND: Apoptosis limits hepatocyte viability in bioartificial livers in vitro and may contribute to liver dysfunction in vivo. Nitric oxide (NO) inhibits hepatocyte apoptosis; however, methods to deliver NO in a sustained manner to hepatocytes are limited. Here, we tested the feasibility of inducible NO synthase (iNOS) gene transfer as an approach to deliver an intracellular source of NO to inhibit spontaneous and tumor necrosis factor-alpha (TNF-alpha)-induced apoptosis in cultured hepatocytes. METHODS: An adenoviral vector carrying the human iNOS gene (AdiNOS) was used to overexpress iNOS in cultured rat hepatocytes. Spontaneous apoptosis was induced by prolonged culture (4 days), and stimulated apoptosis was induced by exposure to TNF-alpha + actinomycin D (TNF-alpha ActD). Nitrite (NO2-), cell viability, and cellular caspase-3-like protease activity were measured. RESULTS: AdiNOS gene transfer resulted in sustained NO production and protected hepatocytes from spontaneous and TNF-alpha + ActD-induced apoptosis. Apoptosis was associated with increases in caspase-3-like protease activity, which was suppressed by iNOS gene transfer in an NO-dependent manner. Dithiothreitol partially reversed the NO-induced suppression of caspase-3-like activity, which is consistent with S-nitrosylation of caspase-3. CONCLUSIONS: Adenovirus-mediated iNOS gene transfer effectively blocks spontaneous and TNF-alpha + ActD-induced cell killing in hepatocytes. iNOS gene transfer could be used to suppress apoptotic hepatocyte death in vitro and possibly in vivo.


Assuntos
Adenoviridae , Apoptose/fisiologia , Caspases , Técnicas de Transferência de Genes , Fígado/citologia , Óxido Nítrico Sintase/genética , Animais , Apoptose/efeitos dos fármacos , Caspase 3 , Cisteína Endopeptidases/metabolismo , Dactinomicina/farmacologia , Ditiotreitol/farmacologia , Precursores Enzimáticos/metabolismo , Humanos , Fígado/enzimologia , Masculino , Óxido Nítrico Sintase Tipo II , Inibidores da Síntese de Proteínas/farmacologia , Ratos , Ratos Sprague-Dawley , Reagentes de Sulfidrila/farmacologia , Fator de Necrose Tumoral alfa/farmacologia
20.
Mol Med ; 4(3): 179-90, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9562976

RESUMO

BACKGROUND: Nitric oxide (NO) is cytostatic for proliferating cells, inhibits microbial growth, and down-regulates the synthesis of specific proteins. Studies were undertaken to determine the mechanism by which NO inhibits total protein synthesis and whether the inhibition correlates with established cytostatic activities of NO. MATERIALS AND METHODS: In in vitro experiments, various cell types were exposed to NO using either donors or expression of inducible NO synthase (iNOS). The capacity of NO to suppress total protein synthesis, measured by incorporation of 35S-methionine into protein, was correlated with the capacity of NO to suppress cell proliferation, viral replication, or iNOS expression. Phosphorylation of eIF-2 alpha was examined as a possible mechanism for the suppressed protein synthesis by NO. RESULTS: Both NO donors and expression of the iNOS suppressed total protein synthesis in L929 cells and A2008 human ovarian tumor cells in parallel with decreased cell proliferation. Suppressed protein synthesis was also shown to correlate with decreased vaccinia virus proliferation in murine peritoneal macrophages in an iNOS-dependent manner. Furthermore, iNOS expression in pancreatic islets or RAW264.7 cells almost completely inhibited total protein synthesis, suggesting that nonspecific inhibition of protein synthesis may be the mechanism by which NO inhibited the synthesis of specific proteins such as insulin or iNOS itself. This possibility was confirmed in RAW264.7 cells where the inhibition of total protein synthesis correlated with the decreased iNOS protein. The decrease in protein levels occurred without changes in iNOS mRNA levels, implicating an inhibition of translation. Mechanistic studies revealed that iNOS expression in RAW264.7 cells resulted in the phosphorylation of eIF-2 alpha and inhibition of the 80S ribosomal complex formation. CONCLUSIONS: These results suggest that NO suppresses protein synthesis by stimulating the phosphorylation of eIF-2 alpha. Furthermore, our observations indicate that nonspecific inhibition of protein synthesis may be a generalized response of cells exposed to high levels of NO and that inhibition of protein synthesis may contribute to many of the described cytostatic actions of NO.


Assuntos
Fator de Iniciação 2 em Eucariotos/metabolismo , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico/fisiologia , Biossíntese de Proteínas , Animais , Divisão Celular , Células Cultivadas , Feminino , Regulação Enzimológica da Expressão Gênica , Humanos , Ilhotas Pancreáticas/enzimologia , Ilhotas Pancreáticas/metabolismo , Células L , Macrófagos Peritoneais/enzimologia , Macrófagos Peritoneais/virologia , Camundongos , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Neoplasias Ovarianas , Penicilamina/análogos & derivados , Penicilamina/farmacologia , Fosforilação , RNA Mensageiro/análise , Ribossomos/metabolismo , S-Nitroso-N-Acetilpenicilamina , Vaccinia virus/fisiologia , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...