Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Am J Transplant ; 12(8): 2125-32, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22548839

RESUMEN

Many potential kidney transplant recipients are unable to receive a live donor transplant due to crossmatch or blood type incompatibility. Kidney paired donation increases access to live donor transplantation but has been significantly underutilized. We established a kidney paired donation program including consented incompatible donor/recipient pairs as well as compatible pairs with older non-human leukocyte antigen identical donors. Over a 3-year period, a total of 134 paired donor transplants were performed, including 117 incompatible pairs and 17 compatible pairs. All transplants were done with negative flow cytometry crossmatches and five were done with desensitization combined with paired donation. Kidney paired donation transplants included two-way and three-way exchanges as well as three chains initiated by nondirected donors. Of the sensitized recipients transplanted by paired donation, 44% had calculated panel reactive antibody levels greater than 80%. Transplantation of females and prior transplant recipients was significantly higher with paired donation. Only three episodes of rejection occurred and no transplants were lost due to rejection. These data highlight the potential of kidney paired donation and suggest that all transplant centers should be actively engaged in paired donation to increase access to live donor transplantation.


Asunto(s)
Trasplante de Riñón , Obtención de Tejidos y Órganos , Prueba de Histocompatibilidad , Humanos , Texas , Resultado del Tratamiento
2.
Diabetes ; 50(3): 515-20, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11246870

RESUMEN

The ability to transfer immunoregulatory, cytoprotective, or antiapoptotic genes into pancreatic islet cells may allow enhanced posttransplantation survival of islet allografts and inhibition of recurrent autoimmune destruction of these cells in type 1 diabetes. However, transient transgene expression and the tendency to induce host inflammatory responses have limited previous gene delivery studies using viral transfer vectors. We demonstrate here that recombinant adeno-associated virus (rAAV) serotype 2, a vector that can overcome these limitations, effectively transduces both human and murine pancreatic islet cells with reporter genes as well as potentially important immunoregulatory cytokine genes (interleukin-4, interleukin-10), although a very high multiplicity of infection (10,000 infectious units/islet equivalent) was required. This requirement was alleviated by switching to rAAV serotype 5, which efficiently transduced islets at a multiplicity of infection of 100. Although adenovirus (Ad) coinfection was required for efficient ex vivo expression at early time points, islets transduced without Ad expressed efficiently when they were transplanted under the renal capsule and allowed to survive in vivo. The rAAV-delivered transgenes did not interfere with islet cell insulin production and were expressed in both beta- and non-beta-cells. We believe rAAV will provide a useful tool to deliver therapeutic genes for modulating immune responses against islet cells and markedly enhance longterm graft survival.


Asunto(s)
Dependovirus/genética , Vectores Genéticos , Islotes Pancreáticos/fisiología , Recombinación Genética , Transducción Genética , Adenoviridae/genética , Animales , Trasplante de Células , Células Cultivadas , Dependovirus/inmunología , Técnicas de Transferencia de Gen , Genes Reporteros , Humanos , Insulina/metabolismo , Interleucina-10/genética , Interleucina-4/genética , Islotes Pancreáticos/citología , Riñón/fisiología , Ratones , Ratones Endogámicos C57BL , Serotipificación
3.
Cell Mol Biol (Noisy-le-grand) ; 51(4): 357-62, 2005 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-16309585

RESUMEN

Transforming growth factor-beta (TGF-beta) is implicated in a variety of kidney diseases where it promotes extracellular matrix (ECM) deposition and pro-inflammatory events, but it also stabilizes and attenuates tissue injury through the activation of cytoprotective proteins, including heme oxygenase-1 (HO-1). HO-1 catalyzes the conversion of heme into carbon monoxide (CO), iron, and biliverdin, which is subsequently converted to bilirubin. The beneficial effects of HO-1 induction include decreasing pro-oxidants (heme), increasing anti-oxidants (biliverdin and bilirubin), and producing a vasodilator with anti-apoptotic and anti-inflammatory properties (CO). The reaction products of HO-1 may also have antifibrogenic properties. The purpose of this study is to explore the effects of HO-1 expression and its reaction products on fibronectin, an ECM protein, in the kidney. The results demonstrate that kidneys of HO-1 knockout mice express significantly more fibronectin protein as compared to heterozygote mice. A potent inducer of HO-1, hemin, significantly decreases fibronectin protein with a concomitant increase in HO-1 protein. Cells expressing HO-1, via TGF-beta1 induction, have reduced fibronectin expression. Bilirubin, a product of the heme oxygenase reaction, attenuates TGF-beta1-mediated increases in fibronectin expression. These results indicate that HO-1 induction and activity may modulate the production of ECM components and suggest a potential role for TGF-beta-mediated HO-1 induction in attenuating renal fibrosis.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Hemo-Oxigenasa 1/metabolismo , Túbulos Renales/efectos de los fármacos , Túbulos Renales/patología , Factor de Crecimiento Transformador beta/farmacología , Animales , Bilirrubina/farmacología , Línea Celular , Inducción Enzimática/efectos de los fármacos , Fibronectinas/biosíntesis , Fibrosis , Hemo-Oxigenasa 1/deficiencia , Hemo-Oxigenasa 1/genética , Túbulos Renales/citología , Túbulos Renales/enzimología , Masculino , Ratones , Ratones Noqueados
4.
Curr Mol Med ; 1(2): 245-58, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11899074

RESUMEN

Most viral gene delivery syslems utilized to date have demonstrated significant limitations in practicality and safety due to the level and duration of recombinant transgene expression as well as their induction of host immunogenicity to vector proteins. Recombinant adeno-associated virus (rAAV) vectors appear to offer a vehicle for safe, long-term therapeutic gene transfer; factors afforded through the propensity of rAAV to establish long-term latency without deleterious effects on the host cell and the relative non-immunogenicity of the virus or viral expressed transgenes. The principal historical limitation of this vector system, efficiency of rAAV-mediated transduction, has recently observed a dramatic increase as the titer, purity, and production capacity of rAAV preparations have improved. In terms of systems that could benefit from such improvements, rAAV gene therapy to enhance solid organ transplantation would appear an obvious choice with islet transplantation forming a promising candidate due to the ability to perform viral transductions ex vivo. Currently, islet transplantation can be used to treat type 1 diabetes yet persisting alloimmune and autoimmune responses represent major obstacles to the clinical success for this procedure. The delivery of transgenes capable of interfering with antigenic recognition and/or cell death [e.g., Fas ligand (FasL), Bcl-2, Bcl-XL] as well as imparting tolerance/immunoregulation [e.g., interleukin(IL)-4, IL-10, transforming growth factor (TGF)-beta], or cytoprotection [e.g., heme oxygenase-1 (HO-1), catalase, manganese superoxide dismutase (MnSOD)] may prevent recurrent type 1 diabetes in islet transplantation and offer a promising form of immunotherapy. Research investigations utilizing such systems may also provide information vital to understanding the immunoregulatory mechanisms critical to the development of both alloimmune and autoimmune islet cell rejection mechanisms and recurrent type 1 diabetes.


Asunto(s)
Dependovirus/genética , Diabetes Mellitus Tipo 1/terapia , Técnicas de Transferencia de Gen , Terapia Genética , Supervivencia de Injerto , Trasplante de Islotes Pancreáticos , Animales , Autoinmunidad , Terapia Combinada , Diabetes Mellitus Tipo 1/inmunología , Genes Reporteros , Vectores Genéticos , Humanos , Transgenes , Trasplante Homólogo
5.
Transplant Proc ; 36(2 Suppl): 25S-32S, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15041303

RESUMEN

Cyclosporine and tacrolimus share the same pharmacodynamic property of activated T-cell suppression via inhibition of calcineurin. The introduction of these drugs to the immunosuppressive repertoire of transplant management has greatly improved the outcomes in organ transplantation and constitutes arguably one of the major breakthroughs in modern medicine. To this date, calcineurin inhibitors are the mainstay of prevention of allograft rejection. The experience gained from the laboratory and clinical use of cyclosporine and tacrolimus has greatly advanced our knowledge about the nature of many aspects of immune response. However, the clinical practice still struggles with the shortcomings of these drugs: the significant inter- and intraindividual variability of their pharmacokinetics, the unpredictability of their pharmacodynamic effects, as well as complexity of interactions with other agents in transplant recipients. This article briefly reviews the pharmacological aspects of calcineurin antagonists as they relate to the mode of action and pharmacokinetics as well as drug interactions and monitoring.


Asunto(s)
Inhibidores de la Calcineurina , Ciclosporina/farmacología , Inmunosupresores/farmacología , Tacrolimus/farmacología , Secuencia de Aminoácidos , Animales , Ciclosporina/química , Ciclosporina/farmacocinética , Ciclosporina/uso terapéutico , Interacciones Farmacológicas , Monitoreo de Drogas/métodos , Humanos , Inmunosupresores/farmacocinética , Inmunosupresores/uso terapéutico , Tacrolimus/química , Tacrolimus/farmacocinética , Tacrolimus/uso terapéutico
6.
Am J Transplant ; 7(5): 1112-20, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17456199

RESUMEN

Interleukin-10 (IL-10) is a pleiotropic cytokine that plays a pivotal role in the regulation of immune responses. Hence, we evaluated the effects of a recombinant adeno-associated viral vector 1 (rAAV1) encoding rat IL-10 (rAAV1-IL-10) in a rat model of kidney allograft rejection. Dark Agouti rat kidneys were transplanted into Wistar-Furth (WF) rats 8 weeks following a single intramuscular administration of either rAAV1-IL-10 or rAAV1-green fluorescence protein (GFP). Isografts (WF-WF) served as an additional experimental control. Both allograft and isograft recipients received daily cyclosporine (10 mg/kg) for 14 days after transplantation. Serum IL-10 levels increased at 8, 12 and 16 weeks following vector administration in rAAV1-IL-10-treated animals, but not in rAAV1-GFP and isograft groups. rAAV1-IL-10 treatment resulted in lower BUN and creatinine levels (p<0.001), as well as increased allograft survival rates from 22% to 90%. Allograft histological abnormalities were significantly attenuated in the rAAV1-IL-10-treated rats compared with those of rAAV1-GFP controls. Serum levels of proinflammatory cytokines such as growth-related oncogene were also significantly higher in the rAAV1-GFP group than in the rAAV1-IL-10 group. These data suggest delivery of IL-10 using a rAAV1 vector improves renal function and prolongs graft survival in a rat model of kidney transplant rejection.


Asunto(s)
Dependovirus/genética , Vectores Genéticos , Supervivencia de Injerto/efectos de los fármacos , Interleucina-10/genética , Interleucina-10/farmacología , Trasplante de Riñón/fisiología , Animales , Nitrógeno de la Urea Sanguínea , Creatinina/metabolismo , Citocinas/sangre , Femenino , Rechazo de Injerto/fisiopatología , Rechazo de Injerto/prevención & control , Supervivencia de Injerto/fisiología , Proteínas Fluorescentes Verdes , Inyecciones Intramusculares , Interleucina-10/sangre , Riñón/efectos de los fármacos , Riñón/patología , Riñón/fisiología , Trasplante de Riñón/patología , Modelos Animales , Ratas , Ratas Endogámicas , Ratas Endogámicas WF , Trasplante Homólogo
7.
Biochem Biophys Res Commun ; 194(2): 654-8, 1993 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-7688219

RESUMEN

A porphyrinic sensor was used to monitor nitric oxide released from cultured endothelial and vascular smooth muscle cells obtained from genetically hypertensive rats and from a normotensive reference strain of rats. Endothelial cell nitric oxide synthase (the constitutive enzyme) was stimulated with bradykinin, and vascular smooth muscle cell nitric oxide synthase (the inducible enzyme) was induced with interleukin-1 beta. Both types of cells from hypertensive rats released less nitric oxide than did cells from normotensive rats. The observed deficient nitric oxide release from endothelial and smooth muscle cells may contribute to the elevated vascular tone and increased cell growth described in hypertension.


Asunto(s)
Aminoácido Oxidorreductasas/metabolismo , Endotelio Vascular/enzimología , Hipertensión/enzimología , Hipertensión/genética , Arterias Mesentéricas/enzimología , Músculo Liso Vascular/enzimología , Aminoácido Oxidorreductasas/biosíntesis , Animales , Aorta Torácica , Técnicas Biosensibles , Bradiquinina/farmacología , Células Cultivadas , Inducción Enzimática , Interleucina-1/farmacología , Óxido Nítrico Sintasa , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY
8.
Biochem Biophys Res Commun ; 193(3): 1076-82, 1993 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-8323533

RESUMEN

Porphyrinic sensors were used for the in situ monitoring of nitric oxide release and diffusion in the endothelial cell, as well as its subsequent diffusion from the endothelial cell through the muscle cells found in the rabbit aorta. The experimental data was compared with that predicted based on Fick's equation for linear diffusion. A time delay of 1.5 s between prediction and experimental concentration of NO due to its chemical reactions was observed at the distance of 100 microns from endothelial cell. About 37% of the NO produced is consumed in chemical reactions in the aorta.


Asunto(s)
Aorta/metabolismo , Técnicas Biosensibles , Endotelio Vascular/metabolismo , Músculo Liso Vascular/metabolismo , Óxido Nítrico/metabolismo , Animales , Membrana Celular/metabolismo , Difusión , Cinética , Masculino , Microquímica , Conejos
9.
J Am Soc Nephrol ; 10(3): 481-91, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10073598

RESUMEN

In this study, a nitric oxide (NO) sensor was used to examine the ability of angiotensin II (AngII), AngIV, and bradykinin (Bk) to stimulate NO release from porcine pulmonary artery (PPAE) and porcine aortic endothelial (PAE) cells and to explore the mechanism of the AngII-stimulated NO release. Physiologic concentrations of AngII, but not Bk, caused release of NO from PPAE cells. In contrast, Bk, but not AngII, stimulated NO release from PAE cells. AngIII-stimulated NO release from PPAE cells required extracellular L-arginine and was inhibited by L-nitro-arginine methyl ester. AT1 and AT2 receptor inhibition had no affect on AngII-mediated NO release or activation of NO synthase (NOS). AngIV, an AngII metabolite with binding sites that are pharmacologically distinct from the classic AngII receptors, stimulated considerably greater NO release and greater endothelial-type constitutive NOS activity than the same amount of AngII. The AngIV receptor antagonist, divalinal AngIV, blocked both AngII- and AngIV-mediated NO release as well as NOS activation. The results demonstrate that AngIV and the AngIV receptor are responsible, at least in part, for AngII-stimulated NO release and the associated endothelium-dependent vasorelaxation. Furthermore, these results suggest that differences exist in both AngII- and Bk-mediated NO release between PPAE and PAE cells, which may reflect important differences in response to these hormones between vascular beds.


Asunto(s)
Angiotensina II/análogos & derivados , Angiotensina II/fisiología , Endotelio Vascular/metabolismo , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico/biosíntesis , Angiotensina II/farmacología , Animales , Bradiquinina/farmacología , Bradiquinina/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Losartán/farmacología , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/análisis , Óxido Nítrico Sintasa/efectos de los fármacos , Arteria Pulmonar/citología , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Valores de Referencia , Porcinos , Vasoconstrictores/farmacología
10.
Gene Ther ; 10(18): 1551-8, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12907946

RESUMEN

Human pancreatic islet cells and hepatocytes represent the two most likely target cells for genetic therapy of type I diabetes. However, limits to the efficiency of rAAV serotype 2 (rAAV2)-mediated gene transfer have been reported for both of these cell targets. Here we report that nonserotype 2 AAV capsids can mediate more efficient transduction of islet cells, with AAV1 being the most efficient serotype in murine islets, suggesting that receptor abundance could be limiting. In order to test this, we generated rAAV particles that display a ligand (ApoE) that targets the low-density lipoprotein receptor, which is present on both of these cell types. The rAAV/ApoE viruses greatly enhanced the efficiency of transduction of both islet cells ex vivo and murine hepatocytes in vivo when compared to native rAAV2 serotype (220- and four-fold, respectively). The use of receptor-targeted rAAV particles may circumvent the lower abundance of receptors on certain nonpermissive cell types.


Asunto(s)
Apolipoproteínas E/genética , Diabetes Mellitus Tipo 1/terapia , Marcación de Gen/métodos , Terapia Genética/métodos , Islotes Pancreáticos/metabolismo , Transducción Genética/métodos , Animales , Células Cultivadas , Dependovirus/genética , Diabetes Mellitus Tipo 1/metabolismo , Vectores Genéticos/administración & dosificación , Humanos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL
11.
J Biol Chem ; 269(44): 27580-8, 1994 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-7525557

RESUMEN

Cellular constituents of heart muscle contain both constitutive and inducible nitric oxide (NO) signaling pathways that modulate the contractile properties of cardiac myocytes. The identities of the inducible NO synthase (iNOS) isoform(s) expressed in cardiac muscle, and of the specific cell types expressing iNOS activity, remain poorly characterized. We amplified a 217-base pair cDNA by reverse transcriptase-polymerase chain reaction from primary cultures of inflammatory cytokine-pretreated adult rat ventricular myocytes (ARVM) that was nearly identical to other iNOS cDNA sequences. Using this 217-base pair cDNA as a probe in Northern blots, we found no evidence of iNOS mRNA in control myocytes, but both interleukin-1 beta and interferon-gamma individually increased iNOS mRNA abundance in primary cultures of ARVM, with maximal expression at 12 h. The half-life of iNOS mRNA in actinomycin C1-treated cells was 4 h. Both dexamethasone and transforming growth factor-beta attenuated the induction of iNOS mRNA abundance and enzyme activity by IL-1 beta and INF gamma. Pretreatment with dexamethasone also abolished the induction of iNOS mRNA, but not the increase in GTP cyclohydrolase mRNA in purified cardiac myocytes from lipopolysaccharide-injected rats. In order to further characterize the specific cell type producing NO, we used a NO-specific porphyrinic/Nafion-coated microsensor to record NO release from a single, isolated ARVM pretreated with IL-1 beta and IFN gamma in L-arginine-depleted medium. NO release could be detected following microinjection of L-arginine in the vicinity of the cell juxtaposed to the NO microsensor, but not following microinjection of D-arginine, and not from ARVM pretreated with L-N-monomethylarginine. Cytokine-pretreated ARVM that had been maintained in L-arginine-depleted medium also exhibited a depressed contractile response to isoproterenol after addition of L-arginine, but not D-arginine. These results indicate that altered contractile function of cardiac myocytes following exposure to specific inflammatory cytokines is due to induction of myocyte iNOS.


Asunto(s)
Aminoácido Oxidorreductasas/biosíntesis , Miocardio/enzimología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células Cultivadas , Clonación Molecular , Citocinas/farmacología , Cartilla de ADN/química , Inducción Enzimática , GTP Ciclohidrolasa/metabolismo , Expresión Génica/efectos de los fármacos , Glucocorticoides/farmacología , Técnicas In Vitro , Lipopolisacáridos/farmacología , Masculino , Datos de Secuencia Molecular , Contracción Miocárdica , Óxido Nítrico Sintasa , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
12.
Proc Natl Acad Sci U S A ; 98(24): 13913-8, 2001 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-11717448

RESUMEN

The development of spontaneous autoimmune diabetes in nonobese diabetic (NOD) mice provides for their use as a model of human type 1 diabetes. To test the feasibility of muscle-directed gene therapy to prevent type 1 diabetes, we developed recombinant adeno-associated virus (rAAV) vectors containing murine cDNAs for immunomodulatory cytokines IL-4 or IL-10. Skeletal muscle transduction of female NOD mice with IL-10, but not IL-4, completely abrogated diabetes. rAAV-IL-10 transduction attenuated the production of insulin autoantibodies, quantitatively reduced pancreatic insulitis, maintained islet insulin content, and altered splenocyte cytokine responses to mitogenic stimulation. The beneficial effects were host specific, as adoptive transfer of splenocytes from rAAV IL-10-treated animals rapidly imparted diabetes in naive hosts, and the cells contained no protective immunomodulatory capacity, as defined through adoptive cotransfer analyses. These results indicate the utility for rAAV, a vector with advantages for therapeutic gene delivery, to transfer immunoregulatory cytokines capable of preventing type 1 diabetes. In addition, these studies provide foundational support for the concept of using immunoregulatory agents delivered by rAAV to modulate a variety of disorders associated with deleterious immune responses, including allergic reactions, transplantation rejection, immunodeficiencies, and autoimmune disorders.


Asunto(s)
Adyuvantes Inmunológicos , Dependovirus , Diabetes Mellitus Tipo 1/prevención & control , Vectores Genéticos , Interleucina-10/genética , Animales , Dependovirus/genética , Diabetes Mellitus Tipo 1/inmunología , Femenino , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Humanos , Interleucina-10/inmunología , Interleucina-4/genética , Interleucina-4/inmunología , Ratones , Ratones Endogámicos NOD , Músculo Esquelético/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda