Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Transplant ; 7(5): 1112-20, 2007 May.
Article in English | MEDLINE | ID: mdl-17456199

ABSTRACT

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.


Subject(s)
Dependovirus/genetics , Genetic Vectors , Graft Survival/drug effects , Interleukin-10/genetics , Interleukin-10/pharmacology , Kidney Transplantation/physiology , Animals , Blood Urea Nitrogen , Creatinine/metabolism , Cytokines/blood , Female , Graft Rejection/physiopathology , Graft Rejection/prevention & control , Graft Survival/physiology , Green Fluorescent Proteins , Injections, Intramuscular , Interleukin-10/blood , Kidney/drug effects , Kidney/pathology , Kidney/physiology , Kidney Transplantation/pathology , Models, Animal , Rats , Rats, Inbred Strains , Rats, Inbred WF , Transplantation, Homologous
2.
Cell Mol Biol (Noisy-le-grand) ; 51(4): 357-62, 2005 Sep 30.
Article in English | MEDLINE | ID: mdl-16309585

ABSTRACT

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.


Subject(s)
Epithelial Cells/drug effects , Epithelial Cells/enzymology , Heme Oxygenase-1/metabolism , Kidney Tubules/drug effects , Kidney Tubules/pathology , Transforming Growth Factor beta/pharmacology , Animals , Bilirubin/pharmacology , Cell Line , Enzyme Induction/drug effects , Fibronectins/biosynthesis , Fibrosis , Heme Oxygenase-1/deficiency , Heme Oxygenase-1/genetics , Kidney Tubules/cytology , Kidney Tubules/enzymology , Male , Mice , Mice, Knockout
3.
Transplant Proc ; 36(2 Suppl): 25S-32S, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15041303

ABSTRACT

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.


Subject(s)
Calcineurin Inhibitors , Cyclosporine/pharmacology , Immunosuppressive Agents/pharmacology , Tacrolimus/pharmacology , Amino Acid Sequence , Animals , Cyclosporine/chemistry , Cyclosporine/pharmacokinetics , Cyclosporine/therapeutic use , Drug Interactions , Drug Monitoring/methods , Humans , Immunosuppressive Agents/pharmacokinetics , Immunosuppressive Agents/therapeutic use , Tacrolimus/chemistry , Tacrolimus/pharmacokinetics , Tacrolimus/therapeutic use
4.
Curr Mol Med ; 1(2): 245-58, 2001 May.
Article in English | MEDLINE | ID: mdl-11899074

ABSTRACT

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.


Subject(s)
Dependovirus/genetics , Diabetes Mellitus, Type 1/therapy , Gene Transfer Techniques , Genetic Therapy , Graft Survival , Islets of Langerhans Transplantation , Animals , Autoimmunity , Combined Modality Therapy , Diabetes Mellitus, Type 1/immunology , Genes, Reporter , Genetic Vectors , Humans , Transgenes , Transplantation, Homologous
5.
J Am Soc Nephrol ; 10(3): 481-91, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10073598

ABSTRACT

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.


Subject(s)
Angiotensin II/analogs & derivatives , Angiotensin II/physiology , Endothelium, Vascular/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/biosynthesis , Angiotensin II/pharmacology , Animals , Bradykinin/pharmacology , Bradykinin/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Losartan/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/analysis , Nitric Oxide Synthase/drug effects , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Reference Values , Swine , Vasoconstrictor Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...