Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 569
Filter
1.
Clin Exp Rheumatol ; 30(1): 73-8, 2012.
Article in English | MEDLINE | ID: mdl-22325451

ABSTRACT

OBJECTIVES: Heme oxygenase-1 (HO-1) which degrades Heme to free iron, biliverdin and carbon monoxide (CO) plays an important role in inflammation. There are, however, conflicting data concerning the role of HO-1 in rheumatoid arthritis (RA) and the therapeutic potential of individual heme degradation products remains to be determined. We therefore investigated the effect of CO and biliverdin upon therapeutic administration in the murine collagen induced arthritis (CIA) model of RA. METHODS: CIA was induced in DBA/1 mice. Anti-CII antibody levels were determined by ELISA. Mice were scored for paw swelling and grip strength. After the first clinical signs of arthritis one group of animals was treated with biliverdin, the second group was treated with CO. After 60 days all animals were sacrificed and analysed for histomorphological signs of arthritis. RESULTS: All animals immunised with CII developed serum anti-CII antibodies. Antibody levels were decreased in the CO-treated group. Both, Biliverdin and the CO-treated animals, showed an improvement in clinical disease activity. Histological analysis revealed significantly less inflammation, erosion and reduced numbers of osteoclasts in CO-treated animals only, whereas cartilage degradation was prevented in both biliverdin and CO-treated animals. CONCLUSIONS: Our data demonstrate a beneficial effect of CO, in particular, and biliverdin, on inflammation and bone destruction in the CIA mouse model.


Subject(s)
Arthritis, Experimental/drug therapy , Biliverdine/therapeutic use , Carbon Monoxide/therapeutic use , Heme Oxygenase-1/metabolism , Joints/drug effects , Administration, Inhalation , Animals , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Biliverdine/administration & dosage , Biliverdine/metabolism , Carbon Monoxide/administration & dosage , Carbon Monoxide/metabolism , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Joints/metabolism , Joints/pathology , Mice
2.
FASEB J ; 19(14): 2045-7, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16223783

ABSTRACT

Endotoxic shock, one of the most prominent causes of mortality in intensive care units, is characterized by pulmonary hypertension, systemic hypotension, heart failure, widespread endothelial activation/injury, and clotting culminating in disseminated intravascular coagulation and multi-organ system failure. In the last few years, studies in rodents have shown that administration of low concentrations of carbon monoxide (CO) exerts potent therapeutic effects in a variety of diseases/disorders. In this study, we have administered CO (one our pretreatment at 250 ppm) in a clinically relevant, well-characterized model of LPS-induced acute lung injury in pigs. Pretreatment only with inhaled CO significantly ameliorated several of the acute pathological changes induced by endotoxic shock. In terms of lung physiology, CO pretreatment corrected the LPS-induced changes in resistance and compliance and improved the derangement in pulmonary gas exchange. In terms of coagulation and inflammation, CO reduced the development of disseminated intravascular coagulation and completely suppressed serum levels of the proinflammatory IL-1beta in response to LPS, while augmenting the anti-inflammatory cytokine IL-10. Moreover, the effects of CO blunted the deterioration of kidney and liver function, suggesting a beneficial effect in terms of end organ damage associated with endotoxic shock. Lastly, CO pretreatment prevents LPS-induced ICAM expression on lung endothelium and inhibits leukocyte marginalization on lung parenchyma.


Subject(s)
Carbon Monoxide/metabolism , Respiration Disorders/prevention & control , Shock, Septic/prevention & control , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis , Blood Coagulation , Carboxyhemoglobin/metabolism , Disease Models, Animal , Heme/chemistry , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1/metabolism , Intercellular Adhesion Molecule-1/metabolism , Interleukin-10/blood , Lipopolysaccharides/chemistry , Lipopolysaccharides/metabolism , Lung/metabolism , Lung/pathology , Models, Biological , Oxygen/metabolism , Swine , Up-Regulation
3.
FASEB J ; 17(12): 1724-6, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12958189

ABSTRACT

Heme oxygenase-1 (HO-1) is induced under a variety of pro-oxidant conditions such as those associated with ischemia-reperfusion injury (IRI) of transplanted organs. HO-1 cleaves the heme porphyrin ring releasing Fe2+, which induces the expression of the Fe2+ sequestering protein ferritin. By limiting the ability of Fe2+ to participate in the generation of free radicals through the Fenton reaction, ferritin acts as an anti-oxidant. We have previously shown that HO-1 protects transplanted organs from IRI. We have linked this protective effect with the anti-apoptotic action of HO-1. Whether the iron-binding properties of ferritin contributed to the protective effect of HO-1 was not clear. We now report that recombinant adenovirus mediated overexpression of the ferritin heavy chain (H-ferritin) gene protects rat livers from IRI and prevents hepatocellular damage upon transplantation into syngeneic recipients. The protective effect of H-ferritin is associated with the inhibition of endothelial cell and hepatocyte apoptosis in vivo. H-ferritin protects cultured endothelial cells from apoptosis induced by a variety of stimuli. These findings unveil the anti-apoptotic function of H-ferritin and suggest that H-ferritin can be used in a therapeutic manner to prevent liver IRI and thus maximize the organ donor pool used for transplantation.


Subject(s)
Apoptosis , Ferritins/genetics , Liver Diseases/prevention & control , Reperfusion Injury/prevention & control , Adenoviridae/genetics , Animals , Cattle , Cytoprotection , Endothelium/cytology , Ferritins/physiology , Genetic Vectors , Liver/metabolism , Liver Diseases/etiology , Liver Diseases/metabolism , Liver Transplantation/adverse effects , Mice , Models, Biological , Rats , Reperfusion Injury/etiology , Reperfusion Injury/metabolism
4.
Transplant Proc ; 35(5): 1798-9, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12962799

ABSTRACT

OBJECTIVE: To explore putative cytoprotective functions of biliverdin during hepatic ischemia/reperfusion (I/R) injury in rat models. MATERIAL AND METHODS: Male Sprague Dawley (SD) rat livers were harvested and stored for 24 hours at 4 degrees C in University of Wisconsin (UW) solution (n=18), and then perfused with blood for 2 hours on an isolated rat liver perfusion apparatus equipped for temperature (37 degrees C), pressure (13 cm H2O), and pH (7.3) maintenance. Biliverdin was added to the blood at concentrations of 10 and 50 micromol in two groups of six animals. Portal vein blood flow, bile production, and GOT/GPT levels were assessed serially. At the conclusion of the experiment, liver samples were collected for histologic evaluation using Suzuki criteria. RESULTS: BV exerted protective effects against liver I/R injury. Adjunctive biliverdin improved portal venous blood flow (mL/min/g) from the beginning of reperfusion (1.33+/-0.17 versus 0.98+/-0.15; P<.001) and increased bile production (mL/g) as compared with the control group (3.40 versus 1.88; P<.003). I/R-induced hepatocellular damage as measured by GOT/GPT release (IU/L) was diminished in the biliverdin group (91 versus 171 and 46 versus 144, respectively; P<.0001). Improved liver function by biliverdin was accompanied by preservation of the histologic structure as assessed by Suzuki criteria (3.7+/-1.4 versus 6.8+/-0.8 in untreated controls; P<.005). CONCLUSIONS: Biliverdin attenuates the ischemia/early reperfusion injury of rat liver grafts as assessed by hemodynamics, function, enzyme analysis, and histology. This study provides the rationale for novel therapeutic approaches using biliverdin to maximize the organ donor pool through the safer use of liver transplants despite prolonged periods of cold ischemia.


Subject(s)
Biliverdine/pharmacology , Liver , Portal System/drug effects , Reperfusion Injury/prevention & control , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Bile/metabolism , Liver/blood supply , Liver/pathology , Male , Perfusion/methods , Rats , Rats, Sprague-Dawley
5.
Gut ; 52(9): 1278-85, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12912858

ABSTRACT

BACKGROUND: Intestinal transplantation provokes an intense inflammatory response within the graft muscularis that causes intestinal ileus. We hypothesised that endogenously produced anti-inflammatory substances could be utilised as novel therapeutics. Therefore, we tested the protective effects of inhaled carbon monoxide (CO) and an endogenous haeme oxygenase 1 (HO-1) anti-inflammatory mediator on transplant induced inflammatory responses and intestinal ileus in the rat. METHODS: Gastrointestinal transit of non-absorbable FITC labelled dextran and in vitro jejunal circular muscle contractions were measured in controls and syngeneic orthotopic transplanted animals with and without CO inhalation (250 ppm for 25 hours). Inflammatory mRNAs for interleukin (IL)-6, IL-1beta, tumour necrosis factor alpha (TNF-alpha), intercellular adhesion molecule 1 (ICAM-1), inducible nitric oxide (iNOS), cyclooxygenase 2 (COX-2), and IL-10 were quantified by real time reverse transcriptase-polymerase chain reaction and HO-1 by northern blot. Histochemical stains characterised neutrophil infiltration and enterocyte apoptosis. RESULTS: Transplantation delayed transit and suppressed jejunal circular muscle contractility. Transplantation induced dysmotility was significantly improved by CO inhalation. Transplantation initiated a significant upregulation in IL-6, IL-1beta, TNF-alpha, ICAM-1, iNOS, COX-2, and HO-1 mRNAs with the graft muscularis. CO inhalation significantly decreased expression of IL-6, IL-1beta, iNOS, and COX-2 mRNAs. CO also significantly decreased serum nitrite levels (iNOS activity). CONCLUSIONS: CO inhalation significantly improved post-transplant motility and attenuated the inflammatory cytokine milieu in the syngeneic rat transplant model. Thus clinically providing CO, the end product of the anti-inflammatory HO-1 pathway, may prove to be an effective therapeutic adjunct for clinical small bowel transplantation.


Subject(s)
Carbon Monoxide/administration & dosage , Gastrointestinal Motility/immunology , Intestine, Small/transplantation , Animals , Bethanechol/pharmacology , Blotting, Northern , Cyclooxygenase 2 , Cytokines/metabolism , Gastrointestinal Motility/drug effects , Gastrointestinal Motility/physiology , Gastrointestinal Transit/immunology , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1 , Inflammation/etiology , Inflammation/physiopathology , Intercellular Adhesion Molecule-1/metabolism , Interleukin-1/metabolism , Interleukin-10/metabolism , Interleukin-6/metabolism , Intestine, Small/immunology , Intestine, Small/physiology , Isoenzymes/metabolism , Male , Muscle Contraction/drug effects , Nitric Oxide Synthase/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/metabolism
7.
J Investig Med ; 49(6): 566-71, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11730093

ABSTRACT

BACKGROUND: Several problems can occur after allogeneic islet transplantation: primary nonfunction, rejection, and the recurrence of autoimmune disease, which involve attack by the recipient's cytokines, T cells, natural killer cells, and monocytes on the donor's beta cells, which leads to beta-cell destruction. Recent studies have revealed that loss of transplanted islets is caused mainly by apoptosis. Heme oxygenase-1 (HO-1) is one of the antiapoptotic genes up-regulated under stress conditions. The aim of this work was to investigate any mechanisms of HO-1-mediated protection of beta cells from apoptosis. METHODS: Apoptosis was assessed by comparison of viable transfected cells with and without apoptotic stimuli, and with and without HO-1 overexpression. Activation and function of p38 mitogen-activated protein kinase were determined using the specific inhibitor SB203580. RESULTS: We have shown that HO-1 mediates antiapoptotic effects in beta cells. The percentage of apoptotic cells after stimulation with tumor necrosis factor a decreased from 75% without HO-1 to 5% when HO-1 was overexpressed. Our data indicate that HO-1 acts as a signal terminator of tumor necrosis factor alpha-induced apoptosis by modulation of the p38 mitogen-activated protein kinase pathway. CONCLUSIONS: Profound cell stress that occurs in islets after transplantation, as well as at the onset of diabetes, results in beta-cell loss through apoptosis. Protection of beta cells by HO-1 improves their survival in vitro after various proapoptotic stimuli, suggesting that HO-1 suppresses one or several signaling pathways leading to apoptosis. We hypothesize that our in vitro findings can be extrapolated to the in vivo situation, and we propose that expression of HO-1 in islets may illuminate a valuable new approach to improving diabetes treatment.


Subject(s)
Apoptosis , Heme Oxygenase (Decyclizing)/physiology , Islets of Langerhans/pathology , Animals , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Heme Oxygenase-1 , Membrane Proteins , Mice , Mitogen-Activated Protein Kinases/physiology , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology , p38 Mitogen-Activated Protein Kinases
8.
Eur Respir J ; 18(2): 420-30, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11529303

ABSTRACT

In recent years, there has been increasing interest in noninvasive monitoring of airway inflammation and oxidative stress. Several volatile and nonvolatile substances can be measured in exhaled breath and have been suggested as potential biomarkers of these events. Exhaled gases, including carbon monoxide (CO), alkanes (ethane, pentane), and substances measured in breath condensate, such as hydrogen peroxide (H2O2) and isoprostanes were all suggested as potential markers of oxidative stress in the lung. A European Respiratory Society (ERS) International Research Seminar entitled "Haemoxygenase-1 induction and exhaled markers of oxidative stress in lung diseases" was organized by the Airway Regulation and Provocation Group of the Clinical Allergy and Immunology Assembly in Budapest, Hungary in September, 1999 to integrate the latest knowledge on these issues and accelerate further improvement in this area. During this 2-day event several issues were raised about: the use and standardization of measurements in exhaled breath; problems of measuring expired H2O2 and other mediators in breath condensate; role and regulation of haemoxygenase (HO)-1 in the lung; and conditions and factors influencing exhaled CO. This report is a summary of the main presentations at the seminar, together with the current areas of research in this rapidly expanding field.


Subject(s)
Heme Oxygenase (Decyclizing)/biosynthesis , Lung Diseases/metabolism , Oxidative Stress , Antioxidants/pharmacology , Biomarkers/analysis , Breath Tests , Carbon Monoxide/metabolism , Enzyme Induction/drug effects , Humans , Lung Diseases/enzymology , Oxidative Stress/drug effects
9.
Diabetes ; 50(9): 1983-91, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11522663

ABSTRACT

Transplantation of islets of Langerhans represents a viable therapeutic approach for the treatment of type 1 diabetes. Unfortunately, transplanted islets are susceptible to allogeneic recognition and rejection, recurrence of autoimmunity, and destruction by local inflammation at the site of implantation. The last of these phenomena might not only result in functional impairment and death of islet cells but could also contribute to amplifying the subsequent specific immune response. Induction of islet cell protection against inflammation could therefore be postulated to be a powerful means to improve overall graft fate. Heme oxygenase-1 (HO-1) has been described as an inducible protein capable of cytoprotection via radical scavenging and apoptosis prevention. The purpose of the present study was to analyze whether HO-1 upregulation in a beta-cell line and in freshly isolated murine islets could result in protection from apoptosis and improve in vivo functional performance. HO-1 upregulation was induced reproducibly with protoporphyrins and was correlated with protection from apoptosis induced in vitro with proinflammatory cytokines or Fas engagement. Furthermore, in vivo HO-1 upregulation resulted in improved islet function in a model of marginal mass islet transplantation in rodents. Strategies aimed at inducing HO-1 upregulation might result in improved success in islet transplantation.


Subject(s)
Apoptosis/physiology , Heme Oxygenase (Decyclizing)/metabolism , Islets of Langerhans Transplantation , Islets of Langerhans/physiology , Animals , Blood Glucose/metabolism , Enzyme Induction , Heme Oxygenase-1 , Islets of Langerhans/drug effects , Islets of Langerhans/enzymology , Male , Membrane Proteins , Mice , Mice, Inbred C57BL , Protoporphyrins/pharmacology , Reference Values , Time Factors , Transplantation, Isogeneic , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/physiology , Up-Regulation
14.
J Immunol ; 166(6): 4185-94, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11238670

ABSTRACT

Mouse-to-rat cardiac transplants survive long term after transient complement depletion by cobra venom factor and T cell immunosuppression by cyclosporin A. Expression of heme oxygenase-1 (HO-1) by the graft vasculature is critical to achieve graft survival. In the present study, we asked whether this protective effect was attributable to the generation of one of the catabolic products of HO-1, carbon monoxide (CO). Our present data suggests that this is the case. Under the same immunosuppressive regimen that allows mouse-to-rat cardiac transplants to survive long term (i.e., cobra venom factor plus cyclosporin A), inhibition of HO-1 activity by tin protoporphyrin, caused graft rejection in 3--7 days. Rejection was associated with widespread platelet sequestration, thrombosis of coronary arterioles, myocardial infarction, and apoptosis of endothelial cells as well as cardiac myocytes. Under inhibition of HO-1 activity by tin protoporphyrin, exogenous CO suppressed graft rejection and restored long-term graft survival. This effect of CO was associated with inhibition of platelet aggregation, thrombosis, myocardial infarction, and apoptosis. We also found that expression of HO-1 by endothelial cells in vitro inhibits platelet aggregation and protects endothelial cells from apoptosis. Both these actions of HO-1 are mediated through the generation of CO. These data suggests that HO-1 suppresses the rejection of mouse-to-rat cardiac transplants through a mechanism that involves the generation of CO. Presumably CO suppresses graft rejection by inhibiting platelet aggregation that facilitates vascular thrombosis and myocardial infarction. Additional mechanisms by which CO overcomes graft rejection may involve its ability to suppress endothelial cell apoptosis.


Subject(s)
Carbon Monoxide/metabolism , Graft Rejection/metabolism , Graft Rejection/prevention & control , Heart Transplantation/immunology , Heme Oxygenase (Decyclizing)/metabolism , Transplantation, Heterologous/immunology , Acute Disease , Animals , Apoptosis/immunology , Carbon Monoxide/administration & dosage , Carbon Monoxide/physiology , Cell Line , Cell Movement/immunology , Endothelium, Vascular/enzymology , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Environmental Exposure , Enzyme Activation/immunology , Graft Rejection/enzymology , Graft Rejection/pathology , Graft Survival/drug effects , Heart Transplantation/pathology , Heme Oxygenase (Decyclizing)/biosynthesis , Heme Oxygenase (Decyclizing)/physiology , Heme Oxygenase-1 , Macrophages/pathology , Male , Membrane Proteins , Mice , Mice, Inbred BALB C , Monocytes/pathology , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/immunology , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Platelet Aggregation/immunology , Rats , Rats, Inbred Lew , Thrombosis/pathology , Thrombosis/prevention & control , Transplantation, Heterologous/pathology , Up-Regulation/immunology
16.
Immunol Rev ; 184: 275-85, 2001 Dec.
Article in English | MEDLINE | ID: mdl-12086318

ABSTRACT

Endothelial cells (EC) play a pivotal role in regulating inflammatory reactions such as those involved in the rejection of transplanted organs. This occurs through the expression of a series of pro- and anti-inflammatory genes that are associated with the activation of these cells. Presumably, the expression of pro-inflammatory genes promotes events that lead to graft rejection, while expression of anti-inflammatory (protective) genes suppresses those events and thus contributes in sustaining graft survival. Understanding how the expression of these genes is regulated and their mechanism of action are important issues for the development of new therapeutic strategies to suppress graft rejection. We have studied this phenomenon using experimental models of transplantation in rats. We discuss here data that supports the concept that grafts can express anti-inflammatory (protective) genes that mitigate inflammatory reactions leading to graft rejection. The data reviewed focus on the role of one of such genes, the stress responsive gene heme oxygenase-1, and of its byproduct carbon monoxide, which can suppress graft rejection and lead to long-term graft survival.


Subject(s)
Graft Rejection/enzymology , Heme Oxygenase (Decyclizing)/immunology , Animals , Apoptosis , Carbon Monoxide , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Graft Rejection/immunology , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase-1 , Humans , Membrane Proteins
17.
J Exp Med ; 192(7): 1015-26, 2000 Oct 02.
Article in English | MEDLINE | ID: mdl-11015442

ABSTRACT

Heme oxygenase 1 (HO-1) inhibits apoptosis by regulating cellular prooxidant iron. We now show that there is an additional mechanism by which HO-1 inhibits apoptosis, namely by generating the gaseous molecule carbon monoxide (CO). Overexpression of HO-1, or induction of HO-1 expression by heme, protects endothelial cells (ECs) from apoptosis. When HO-1 enzymatic activity is blocked by tin protoporphyrin (SnPPIX) or the action of CO is inhibited by hemoglobin (Hb), HO-1 no longer prevents EC apoptosis while these reagents do not affect the antiapoptotic action of bcl-2. Exposure of ECs to exogenous CO, under inhibition of HO-1 activity by SnPPIX, substitutes HO-1 in preventing EC apoptosis. The mechanism of action of HO-1/CO is dependent on the activation of the p38 mitogen-activated protein kinase (MAPK) signaling transduction pathway. Expression of HO-1 or exposure of ECs to exogenous CO enhanced p38 MAPK activation by TNF-alpha. Specific inhibition of p38 MAPK activation by the pyridinyl imidazol SB203580 or through overexpression of a p38 MAPK dominant negative mutant abrogated the antiapoptotic effect of HO-1. Taken together, these data demonstrate that the antiapoptotic effect of HO-1 in ECs is mediated by CO and more specifically via the activation of p38 MAPK by CO.


Subject(s)
Apoptosis , Carbon Monoxide/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Animals , Cattle , Cell Line , Cells, Cultured , Cyclic GMP/metabolism , Endothelium, Vascular/cytology , Enzyme Activation , Gene Expression , Guanylate Cyclase/metabolism , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/physiology , Heme Oxygenase-1 , Humans , Iron/metabolism , Membrane Proteins , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases
18.
Transplantation ; 70(6): 864-70, 2000 Sep 27.
Article in English | MEDLINE | ID: mdl-11014639

ABSTRACT

BACKGROUND: Extracellular ATP and ADP may be important mediators of vascular inflammation and thrombosis. Nucleoside triphosphate diphosphohydrolase (NTPDase or CD39) is a vascular ectoenzyme that hydrolyses ATP and ADP; however, this activity is lost during reperfusion injury. We show that the supplementation of NTPDase activity within xenograft vasculature using CD39 recombinant adenoviruses (AdCD39) has protective effects in vivo. METHODS: Recombinant adenoviruses containing human CD39 or beta-galactosidase (Adbeta-gal) encoding genes were constructed. Hartley guinea pig coronary arteries were perfused ex vivo with University of Wisconsin solution containing 10(9) plaque-forming units of the recombinant adenovirus. Infected grafts were then implanted in the abdomen of complement depleted Lewis rats. RESULTS: NTPDase activities decreased in all grafts within the first 24 hr and subsequently recovered only in those hearts infected with AdCD39. Immunohistological examination of AdCD39-infected grafts confirmed successful CD39 gene transfer into the endocardium and macrovasculature. Expression of CD39 modestly prolonged graft survival (90.2+/-5.4 hr, mean+/-SD, n=5) when compared with Adbeta-gal-infected grafts (67.4+/-5.4 hr, P<0.005) and perfusion controls (66.4+/-5.2 hr; P<0.005). CONCLUSIONS: Recombinant adenoviral infection can induce expression of CD39 within cardiac xenografts and provide survival benefits in vivo. Our data show that ex vivo infection by recombinant adenovirus vectors can result in vascular expression of a potential therapeutic agent.


Subject(s)
Adenosine Triphosphatases , Adenoviridae/genetics , Antigens, CD/genetics , Genetic Vectors/immunology , Heart Transplantation/immunology , Transplantation, Heterologous/immunology , Animals , Apyrase/metabolism , Blotting, Western , Gene Transfer Techniques , Graft Survival/genetics , Guinea Pigs , Humans , Kinetics , Male , Rats , Transplantation, Heterologous/pathology
20.
J Immunol ; 164(9): 4883-92, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10779798

ABSTRACT

We transplanted hamster hearts into rats that had been sensitized to hamster cardiac grafts 5 days earlier as a model for discordant xenotransplantation. Sensitized rats had high serum levels of elicited anti-donor IgM and IgG that caused hyperacute rejection. Transient complement inhibition with cobra venom factor (CVF) plus daily and continuing cyclosporin A (CyA) prevented hyperacute rejection. However, grafts underwent delayed xenograft rejection (DXR). DXR involved IgG and associated Ab-dependent cell-mediated rejection, because depletion of IgG or Ab-dependent cell-mediated rejection-associated effector cells prolonged graft survival and the serum-mediated Ab-dependent cell-mediated cytotoxicity in vitro. Blood exchange in combination with CVF/CyA treatment dramatically decreased the level of preexisting Abs, but DXR still occurred in association with the return of Abs. Splenectomy and cyclophosphamide acted synergistically to delay Ab return, and when combined with blood exchange/CVF/CyA facilitated long-term survival of grafts. These grafts survived in the presence of anti-donor IgM, IgG, and complement that precipitated rejection of naive hearts, indicating that accommodation (survival in the presence of anti-graft Abs and complement) had occurred. We attribute the long-term survival to the removal of preexisting anti-donor Abs and therapy that attenuated the rate of Ab return. Under such conditions, the surviving hearts showed expression in endothelial cells and smooth muscle cells of protective genes and an intragraft Th2 immune response. Th2 responses and protective genes are associated with resistance to IgM- and IgG-mediated, complement-dependent and -independent forms of rejection.


Subject(s)
Graft Enhancement, Immunologic , Graft Survival/immunology , Heart Transplantation/immunology , Immunization , Transplantation, Heterologous , Animals , Antibodies, Heterophile/biosynthesis , Antibody-Dependent Cell Cytotoxicity , Complement Activation , Cricetinae , Cyclosporine/pharmacology , Cytotoxicity, Immunologic , Exchange Transfusion, Whole Blood , Graft Enhancement, Immunologic/methods , Graft Rejection/immunology , Graft Rejection/pathology , Graft Survival/drug effects , Heart Transplantation/pathology , Immunization/methods , Immunoglobulin G/biosynthesis , Immunoglobulin G/blood , Immunoglobulin G/classification , Immunohistochemistry , Immunosuppression Therapy/methods , Killer Cells, Natural/immunology , Macrophages/immunology , Mesocricetus , Monocytes/immunology , Rats , Rats, Inbred Lew , Transplantation, Heterologous/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...