Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Circulation ; 102(19 Suppl 3): III237-42, 2000 Nov 07.
Article in English | MEDLINE | ID: mdl-11082394

ABSTRACT

BACKGROUND: The long-term success of cardiac transplantation is limited by graft coronary artery disease (GCAD). Antisense oligonucleotides (ASs) to proliferating cell nuclear antigen (PCNA) and Cdc2 kinase (Cdc2 k) can arrest cell cycle progression and inhibit neointimal hyperplasia. Transforming growth factor-ss(1) (TGF-ss(1)) has been implicated in vascular smooth muscle cell (VSMC) activation. The role of TGF-ss(1) in GCAD remains unclear. We hypothesized that ASs to PCNA and Cdc2 k would inhibit VSMC proliferation and GCAD. METHODS AND RESULTS: In vitro VSMC proliferation was determined after pretreatment with AS solution or medium alone followed by angiotensin II stimulation. PVG-to-ACI rat heterotopic cardiac transplantation procedures were performed after ex vivo pressure-mediated transfection of ASs to PCNA and Cdc2k or saline alone. At postoperative days 30, 60, and 90, allografts were assessed for GCAD, percent neointimal macrophages and VSMCs, and TGF-ss(1) activity. AS pretreatment significantly attenuated VSMC proliferation. At postoperative day 90, percent affected arteries, percent occlusion, and intima-media ratio demonstrated severe GCAD in saline-treated allografts, whereas these parameters were significantly lower in AS-treated allografts. Percent neointimal macrophages and VSMCs was reduced in AS-treated allografts. TGF-ss(1) activity was increased in saline compared with AS-treated allografts and nontransplanted heart controls. CONCLUSIONS: ASs to PCNA and Cdc2 k inhibit VSMC proliferation in vitro and reduce GCAD, percent neointimal VSMCs and macrophages, and TGF-ss(1) activity in vivo. TGF-ss(1) may play a "response to injury" role in the development of GCAD. The prevention of GCAD via AS inhibition of cell cycle regulatory genes before reperfusion may offer a useful clinical alternative to current therapeutic strategies.


Subject(s)
CDC2 Protein Kinase/antagonists & inhibitors , Coronary Disease/prevention & control , Heart Transplantation/adverse effects , Oligonucleotides, Antisense/therapeutic use , Proliferating Cell Nuclear Antigen/metabolism , Actins/metabolism , Animals , CDC2 Protein Kinase/genetics , Cell Division/drug effects , Cells, Cultured , Coronary Disease/diet therapy , Coronary Disease/etiology , Coronary Disease/metabolism , Coronary Disease/pathology , Disease Models, Animal , Humans , Immunohistochemistry , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Polymerase Chain Reaction , Proliferating Cell Nuclear Antigen/genetics , Rats , Rats, Inbred ACI , Rats, Sprague-Dawley , Tetrazolium Salts , Thiazoles , Transcription, Genetic/drug effects , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1
2.
Circulation ; 100(1): 67-74, 1999 Jul 06.
Article in English | MEDLINE | ID: mdl-10393683

ABSTRACT

BACKGROUND: Chronic graft vascular disease (CGVD) in cardiac allografts has been defined as a slowly evolving vasculopathy unresponsive to conventional immunosuppression. We compared 4 rodent models of CGVD to evaluate the reproducibility of CGVD in heart allografts. Rapamycin (Rapa) and cyclosporine (CSA) were then used to treat CGVD. METHODS AND RESULTS: Hearts were harvested and placed heterotopically into allogenic recipients. CGVD scores of PVG allografts from ACI recipients treated with CSA on days 1 through 10 were significantly elevated on day 90 (n=16) compared with other models (immunosuppression used): (1) Lewis to F344 recipients (CSA), (2) Brown Norway to Lewis (FK506), and (3) DA to Wistar-Firth (methylprednisolone, azathioprine, CSA). Although delayed (day 60 to 90) CSA treatment had no effect (n=6), delayed Rapa (3 mg. kg-1. d-1 IP) reversed CGVD in PVG grafts (0.22+/-0.19 on day 90, n=6). ACI isografts showed no evidence of CGVD (n=6) at day 90. Immunohistochemistry of PVG grafts revealed perivascular infiltrates consisting of CD4(+) T cells and limited numbers of macrophages persisting up to day 90. Flow cytometry demonstrated increased levels of anti-donor antibody at day 90, which was significantly inhibited by Rapa treatment. CONCLUSIONS: PVG grafts developed a significant increase in CGVD without evidence of ongoing myocardial rejection. This CGVD appeared to be mediated by both cellular and humoral mechanisms, given CD4(+) perivascular infiltrates and increased levels of anti-donor antibody. The anti-CGVD effectiveness of Rapa during a period in which there was little myocardial cellular infiltrate supports a novel mechanism of effect such as smooth muscle or B-cell inhibition.


Subject(s)
Coronary Disease/drug therapy , Graft vs Host Disease/drug therapy , Heart Transplantation/adverse effects , Immunosuppressive Agents/therapeutic use , Sirolimus/therapeutic use , Animals , Antibody Formation/drug effects , Antibody Specificity , Coronary Disease/etiology , Coronary Disease/immunology , Cyclosporine/therapeutic use , Drug Evaluation, Preclinical , Flow Cytometry , Graft Rejection , Graft vs Host Disease/etiology , Heart Transplantation/immunology , Histocompatibility , Histocompatibility Antigens/immunology , Immunity, Cellular/drug effects , Immunoglobulin G/blood , Isoantibodies/blood , Male , Nitric Oxide/physiology , Rats , Rats, Inbred ACI , Rats, Inbred F344 , Rats, Inbred Lew , Rats, Inbred WF , Reproducibility of Results , Transplantation, Homologous/adverse effects , Transplantation, Homologous/immunology
3.
Circulation ; 104(12 Suppl 1): I202-6, 2001 Sep 18.
Article in English | MEDLINE | ID: mdl-11568056

ABSTRACT

BACKGROUND: Oxidative stress after ischemia/reperfusion of cardiac allografts leads to cytokine production. Bcl-2, an inhibitor of apoptosis, also has strong antioxidant properties. Caspase-3 is known to cleave bcl-2. This study tests the hypothesis that bcl-2 is downregulated while tumor necrosis factor-alpha (TNF-alpha) levels increase after cardiac transplantation. Furthermore, the use of caspase-3 inhibition was investigated as a strategy for preserving myocardial bcl-2 and mitochondrial cytochrome c after transplantation. METHODS AND RESULTS: PVG-to-ACI rat heterotopic cardiac transplantations were performed in 4 groups designed with 30 minutes' ischemia and 4 or 8 hours of reperfusion (n=4 per group). Treatment consisted of DEVD-CHO 500 microgram IP per animal to donor and recipient 2 hours before transplantation and 250 microgram IC into allograft. Controls were treated with saline. Grafts were analyzed by reverse transcription-polymerase chain reaction for bcl-2 mRNA, by ELISA for TNF-alpha, for myeloperoxidase activity, and by Western blot for cytochrome c. In untreated groups, bcl-2 mRNA decreased significantly over time, whereas TNF-alpha increased significantly at 4 hours (P=0.003) and returned to baseline after 8 hours' reperfusion (P=NS compared with normal hearts). Treatment with caspase-3 inhibitor showed significant upregulation of bcl-2 mRNA expression after 4 and 8 hours of reperfusion (P<0.001 versus control), with a concomitant decrease in TNF-alpha to baseline levels. Myeloperoxidase activity in all groups was no different from that of normal hearts. Mitochondrial cytochrome c release increased in both control and treatment groups. CONCLUSIONS: Bcl-2 is actively downregulated and TNF-alpha is upregulated in this model of cardiac allograft ischemia/reperfusion. Furthermore, the caspase-3 pathway is linked to this process, and blockade of caspase-3 can ameliorate reperfusion injury by upregulating bcl-2 and inhibiting TNF-alpha without affecting cytochrome c release.


Subject(s)
Caspase Inhibitors , Heart Transplantation , Proto-Oncogene Proteins c-bcl-2/metabolism , Reperfusion Injury/prevention & control , Up-Regulation/drug effects , Animals , Caspase 3 , Caspases/metabolism , Cytochrome c Group/metabolism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Heart Transplantation/adverse effects , Male , Mitochondria/enzymology , Myocardium/metabolism , Oligopeptides/pharmacology , Oxidative Stress , Peroxidase/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains , Reperfusion Injury/etiology , Reperfusion Injury/metabolism , Transplantation, Homologous , Tumor Necrosis Factor-alpha/metabolism
4.
Transplantation ; 44(1): 5-8, 1987 Jul.
Article in English | MEDLINE | ID: mdl-3111038

ABSTRACT

The ability of a new cyclosporine (Cs) derivative, (Nva2)-Cs (CsG), to suppress rejection of lung and heart allografts in rats was determined and compared with that of CsA. Left lungs were transplanted orthotopically; hearts were transplanted heterotopically into the abdomen. (Nva2)-Cs was used in three experimental protocols: (1) single or three (Nva2)-Cs injections given to lung-transplanted rats, (2) daily oral (Nva2)-Cs treatment at different doses compared with similar CsA treatments in heart allografted rats, and (3) An 11-day (Nva2)-Cs treatment starting at increasing intervals after transplantation of hearts. (Nva2)-Cs was found to be immunosuppressive, and effective even when the treatment started as late as four days after transplantation. However, (Nva2)-Cs was less effective than CsA in suppressing rejection of lung and heart allografts at low doses. Because (Nva2)-Cs is possibly not nephrotoxic, it might be a useful drug if used in higher doses than CsA or in combination with other immunosuppressive agents.


Subject(s)
Cyclosporine , Cyclosporins/therapeutic use , Graft Rejection/drug effects , Heart Transplantation , Heart-Lung Transplantation , Lung Transplantation , Animals , Cyclosporins/blood , Drug Evaluation, Preclinical , Male , Rats , Rats, Inbred ACI/immunology , Rats, Inbred BN/immunology , Rats, Inbred Lew/immunology
5.
Transplantation ; 70(11): 1560-8, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11152216

ABSTRACT

BACKGROUND: Nuclear factor-kappaB (NF-kappaB) is a transcription factor that upregulates adhesion molecules ICAM-1, VCAM-1, and ELAM-1. We hypothesized the use of ex vivo pressure-mediated delivery of transcription factor decoys (TFD) to NF-kappaB binding sites would decrease expression of adhesion molecules, and decrease reperfusion injury, acute rejection, and graft coronary artery disease (GCAD) in rat cardiac allografts. METHODS: Heterotopic heart transplants were performed on donor hearts treated with saline, 10 mg/kg LPS, 160 micromol/L NF-kappaB TFD, or 160 micromol/L scrambled sequence (NF-SC) TFD for 45 min at 78 psi (6 atm). Transfection efficiency was determined with FITC-labeled TFD. Reverse transcription-PCR and immunohistochemistry was used to analyze adhesion molecule mRNA and protein expression, respectively. Apoptosis was measured with DNA fragmentation analysis. Reperfusion injury was assessed with cardiac edema, neutrophil infiltration, and histology. Acute rejection was determined by daily palpation. Allografts were assessed at POD 90 for the development of GCAD by computer-assisted image analysis to determine intimal:medial ratio and myointimal proliferation. RESULTS: Hyperbaric pressure was an effective method of NF-kappaB TFD delivery (P<0.001 vs. controls). NF-kappaB TFD treatment led to decreased mRNA and protein expression of adhesion molecules. Treatment with NF-kappaB TFD led to a significant decrease in all reperfusion injury parameters compared to saline and NF-SC controls (P<0.01 vs. controls). Higher levels of apoptosis were seen in allografts treated with NF-kappaB TFD compared to control allografts. NF-kappaB TFD treatment prolonged allograft survival over saline and NF-SC controls (P<0.05). Myointimal proliferation and intimal:medial ratios in NF-kappaB TFD-treated allografts were significantly decreased compared to saline and NF-SC treatment (P<0.00001). CONCLUSIONS: Ex vivo pressure-mediated delivery of NF-kappaB TFD is an effective method to block adhesion molcule expression and reperfusion injury in the immediate posttransplant period. Further, NF-kappaB TFD treatment prolongs allograft survival and decreases GCAD.


Subject(s)
Coronary Disease/prevention & control , Heart Transplantation , NF-kappa B/therapeutic use , Transcription Factors/therapeutic use , Animals , Coronary Disease/etiology , Graft Survival/drug effects , Heart Transplantation/adverse effects , Heart Transplantation/immunology , Immunohistochemistry , Male , Oligoribonucleotides, Antisense/pharmacology , Rats , Rats, Inbred ACI , Rats, Sprague-Dawley , Reperfusion Injury/prevention & control , Reverse Transcriptase Polymerase Chain Reaction
6.
Transplantation ; 57(12): 1795-8, 1994 Jun 27.
Article in English | MEDLINE | ID: mdl-8016886

ABSTRACT

With the increasing frequency of transplantation of two or more organs into a single recipient, it has become evident that different organs are rejected with different kinetics. In this study the kinetics of skin, lung, and heart allograft rejection were compared in a rodent model. To study the influence of different allografts on the recipient's immune system, simultaneous or sequential skin, lung, or heart transplants were performed in various combinations, using DA rats as recipients for PVG allografts. Recipients receiving primary allografts were treated postoperatively with ten doses of cyclosporine (CsA) or preoperatively with 4 doses of rabbit antirat thymocyte globulin (ATG). Subsequent transplants were performed a minimum of 40 days later without additional immunosuppression. All primary skin allografts and 60% of primary lung allografts were rejected, while 100% of the heart allografts were accepted indefinitely. Recipients of primary skin allografts rejected subsequent skin, lung, or heart allografts with accelerated kinetics. Recipients of primary heart allografts accepted subsequent skin, lung, and heart allografts indefinitely without further immunosuppression. Surprisingly, animals that had rejected a primary lung allograft accepted subsequent skin or heart allografts indefinitely. Simultaneously transplanted skin and lung allografts were concordantly rejected. However, these animals accepted a subsequent heart allograft indefinitely, suggesting a strong tolerizing effect of lung allografts. Our results indicate that tissue-specific differences are critical, not only in determining acceptance or rejection of a primary allograft but also in determining the fate of subsequent allografts.


Subject(s)
Cyclosporine/pharmacology , Graft Rejection/immunology , Graft Survival/immunology , Heart Transplantation/immunology , Immune Tolerance , Lung Transplantation/immunology , Skin Transplantation/immunology , Animals , Antilymphocyte Serum/pharmacology , Graft Survival/drug effects , Male , Rats , Rats, Inbred Lew , Rats, Inbred Strains , Time Factors , Transplantation, Homologous/immunology
7.
Transplantation ; 59(10): 1444-52, 1995 May 27.
Article in English | MEDLINE | ID: mdl-7539554

ABSTRACT

Cellular mechanisms responsible for maintenance of peripheral transplant tolerance in a rodent model were evaluated. Donor-specific tolerance was established in ACI rats given a vascularized heterotopic cardiac allograft followed by a 10-day course of cyclosporine. Tolerance was associated with a reduction in donor-specific cytotoxic T lymphocyte precursors and the presence within the spleen of cells capable of transferring suppression in adoptive transfer assays. Experiments using thymectomized animals revealed that the establishment and maintenance of tolerance occurred peripherally, independently of the thymus. Adoptive transfer experiments demonstrated that ongoing graft tolerance was mediated by suppressor cells that were antigen-restricted, radiosensitive, and capable of preventing allograft rejection by naive as well as sensitized cells in vivo. Studies designed to disrupt tolerance demonstrated a remarkable durability of graft protection once established, and give insight into the identity and mechanism of action of suppressor cells generated in this model.


Subject(s)
Cyclosporine/therapeutic use , Heart Transplantation/immunology , Skin Transplantation/immunology , Animals , Antilymphocyte Serum/therapeutic use , Blood Component Transfusion , Epitopes/radiation effects , Graft Rejection/radiotherapy , Graft Survival/drug effects , Immune Tolerance/drug effects , Interleukin-2/pharmacology , Lymphoid Tissue/radiation effects , Male , Radio Waves , Rats , Rats, Inbred ACI , Rats, Inbred Lew , Spleen/cytology , Stem Cells , T-Lymphocytes, Cytotoxic , T-Lymphocytes, Regulatory/immunology
8.
Transplantation ; 68(6): 825-32, 1999 Sep 27.
Article in English | MEDLINE | ID: mdl-10515383

ABSTRACT

BACKGROUND: We hypothesized that ex vivo donor allograft transfection with antisense oligodeoxynucleotide (AS ODN) would inhibit the expression of intercellular adhesion molecule (ICAM)-1, an important mediator of T-cell adhesion and costimulation, and therefore suppress acute cardiac rejection. METHODS: Hearts were transfected ex vivo with AS, reverse AS ODN, or saline by applying 3 atm pressure for 45 min at 4 degrees C. Grafts were then transplanted into allogenic recipients +/- treatment with leukocyte function-associated antigen (LFA)-1 monoclonal antibody (mAb) (1.5 mg/kg intravenously), cyclosporine (2.5 mg/ kg/day p.o.), or rapamycin (0.025 mg/kg/day intraperitoneally). Reperfusion injury was assessed in grafts harvested at early time points using the myeloperoxidase, %wet weight, and %contraction band necrosis assays; transfection efficiency was assessed using fluorescent microscopy; and efficacy of ICAM-1 blockade was assessed using immunohistochemistry. Other grafts were followed until rejection with donor/third-party skin grafting, adoptive transfer, and interleukin 2 infusion studies in selected recipients. RESULTS: Transfection was highly efficient (fluorescein isothiocyanate-ODN in 48+/-5% of total myocardial nuclei), nontoxic, and reduced the ICAM-1-positive area to 53+/-14% versus having no effect on MHC class I expression (n=4). The incidence of survival >60 days after AS ODN + LFA-1 monoclonal antibody was 75%, significantly higher than other regimens. CONCLUSION: AS ODN hyperbaric transfection proved highly efficient, effective at ICAM-1 blockade, and induced cardiac allograft tolerance when combined with LFA-1 monoclonal antibody. This highly targeted alteration of allograft immunogenicity may have an important role in future immunosuppressive strategies.


Subject(s)
Graft Rejection/prevention & control , Heart Transplantation/immunology , Oligodeoxyribonucleotides, Antisense/therapeutic use , Transfection/methods , Acute Disease , Animals , Antibodies, Monoclonal/physiology , Cyclosporine/pharmacology , Genetic Therapy/methods , Graft Survival/drug effects , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/physiology , Lymphocyte Function-Associated Antigen-1/immunology , Male , Rats , Rats, Inbred ACI , Rats, Inbred BN , Rats, Sprague-Dawley , Therapeutic Equivalency , Transplantation, Homologous/immunology
9.
Transplantation ; 69(6): 1067-74, 2000 Mar 27.
Article in English | MEDLINE | ID: mdl-10762209

ABSTRACT

BACKGROUND: Our purpose was to optimize hyperbaric pressure as a vector for ex vivo transfection of antisense oligodeoxynucleotides (AS-ODN) to intercellular adhesion molecule-1 to limit reperfusion injury (RI) in cardiac allografts. We investigated the effects of increased pressure, incubation time, and AS-ODN concentrations on transfection efficiency and toxicity. METHODS AND RESULTS: PVG (RT1c) donor hearts were heterotopically transplanted to ACI (RT1a) recipients. Donor hearts were harvested and the various groups were treated at: (1) different pressure (1-9 atm) for 45 min with 80 micromol/liter AS-ODN; (2) different incubation times (15 min to 6 hr) at 5 atm with 80 micromol/liter AS-ODN; 3) different AS-ODN concentrations (80-240 micromol/liter) at 5 atm for 45 min. Hearts were procured 24 or 72 hr after transplantation. Transfection efficiency was determined with fluorescein-labeled AS-ODN. The degree of RI was determined with biochemical and histological analysis. Increasing pressure from ambient (1 atm) pressure to pressures as high as 9 atm leads to a increase in transfection efficiency from 1.7+/-.5 to 62+/-3.9% and a reduction in RI. Increased incubation time up to 45 min increased transfection efficiency and reduced RI, but longer incubation times induced significant toxicity to the allograft. Increased AS-ODN concentrations improved transfection and reduced RI. CONCLUSIONS: Hyperbaric pressure is a safe and effective vector for the ex vivo delivery of AS-ICAM-1-ODN to rodent cardiac allografts and results in a reduction in reperfusion injury.


Subject(s)
Heart Transplantation , Hyperbaric Oxygenation , Intercellular Adhesion Molecule-1/genetics , Oligonucleotides, Antisense/toxicity , Reperfusion Injury/prevention & control , Reperfusion Injury/therapy , Animals , Genetic Therapy , Male , Oligonucleotides, Antisense/therapeutic use , Pressure , RNA, Messenger/metabolism , Rats , Rats, Inbred ACI , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Time Factors , Transfection
10.
Transplantation ; 50(3): 366-73, 1990 Sep.
Article in English | MEDLINE | ID: mdl-1976282

ABSTRACT

In the present report a monoclonal antibody designated OX-38 directed against the rat CD4 molecule was tested for its ability to prolong the survival of heterotopic vascularized rat heart allografts transplanted across major histocompatibility barriers. Fluorescence-activated cell-sorter analysis showed that administration of OX-38 selectively depleted 80-95% of CD4+ cells from peripheral blood of treated rats. The immunosuppressive effects of OX-38 in vivo were verified by suppression of an antibody response against OX-38 itself as a heterologous protein immunogen. Recipient rats received OX-38 antibody as a single agent given in pretransplant regimens. Nine of 12 treated rats have maintained heterotopic abdominal heart allografts for greater than 175 days. Control rats that did not receive antibody therapy rejected their grafts within 14 days. Rats that maintained heart allografts for greater than 100 days accepted second donor strain hearts but rejected third-party heart grafts transplanted into the femoral space. Anti-CD4-induced allograft unresponsiveness persisted for at least 90 days following surgical removal of donor tissue and retransplantation of a second donor-matched heart. These results indicated that transient, pretransplant therapy with monoclonal antibodies directed against the CD4+ lymphocyte induced specific, long-lasting unresponsiveness to fully MHC-mismatched cardiac allografts in rats without additional immunosuppression.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD4 Antigens/immunology , Graft Survival/immunology , Heart Transplantation/immunology , Animals , Antibody Formation/immunology , Antigens, Differentiation, T-Lymphocyte/physiology , CD4-Positive T-Lymphocytes/immunology , CD8 Antigens , Histocompatibility Antigens/immunology , Immunosuppression Therapy , Lymphocyte Depletion , Male , Rats , Rats, Inbred ACI , Rats, Inbred BN , Rats, Inbred Lew , Transplantation, Homologous
11.
J Thorac Cardiovasc Surg ; 116(3): 386-96, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9731779

ABSTRACT

OBJECTIVE: We hypothesized that ex vivo hyperbaric transfection of antisense oligodeoxynucleotides for blockade of intercellular adhesion molecule-1, an important mediator of cell adhesion and T-cell co-stimulation, would reduce chronic graft vascular disease in cardiac allografts. METHODS: PVG hearts underwent ex vivo transfection with antisense, reverse antisense intercellular adhesion molecule-1 oligodeoxynucleotide (80 micromol/L), or saline solution at 3 atm pressure for 45 minutes at 4 degrees C and were transplanted heterotopically into ACI recipients with or without treatment with intercellular adhesion molecule-1 (1A29) or leukocyte function associated antigen-1 (WT.1) monoclonal antibodies. Transfection efficiency was confirmed with fluorescein isothiocyanate-labeled oligodeoxynucleotides and fluorescent microscopy. Efficacy of intracellular adhesion molecule-1 blockade was assessed with the use of immunohistochemistry. Graft reperfusion injury was evaluated at 6 to 24 hours by neutrophil infiltration (myeloperoxidase [MPO]), cardiac edema (%wt/wt), and histologic injury (percent contraction band necrosis). Grafts from recipients treated with cyclosporine A (5 mg/kg per day, days 0 to 9) were scored for chronic graft vascular disease on postoperative day 90 ranging from 0 (no involvement) to 4 (>50% vascular occlusion). RESULTS: Transfection was highly efficient (fluorescein isothiocyanate-labeled oligodeoxynucleotides in 48%+/-5% of total myocardial nuclei) and effective at blocking intracellular adhesion molecule-1 expression (positive area in allografts taken on postoperative day 3 was reduced from 100%+/-0% to 52%+/-14%, n=4). Blockade with antisense oligodeoxynucleotides versus monoclonal antibodies was less effective at preventing reperfusion injury while more effective at reducing chronic graft vascular disease (score 0.98+/-0.48, p < 0.05). Reverse antisense oligodeoxynucleotides and vector control (antisense oligodeoxynucleotide infusion without pressure) groups failed to demonstrate this beneficial effect. CONCLUSION: Hyperbaric transfection of antisense oligodeoxynucleotides proved highly efficient, effective at blockade of intracellular adhesion molecule-1, and demonstrated a sequence-specific reduction in chronic graft vascular disease. This highly targeted alteration of donor organ immunogenicity may have an important future role in clinical immunosuppressive strategies.


Subject(s)
Coronary Disease/prevention & control , Genetic Therapy/methods , Heart Transplantation , Intercellular Adhesion Molecule-1/genetics , Myocardial Reperfusion Injury/prevention & control , Postoperative Complications/prevention & control , Transfection , Animals , Coronary Disease/etiology , Coronary Disease/immunology , Graft Rejection/immunology , Graft Rejection/prevention & control , Heart Transplantation/adverse effects , Immunosuppression Therapy/methods , Intercellular Adhesion Molecule-1/physiology , Male , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/immunology , Oligonucleotides, Antisense/therapeutic use , Postoperative Complications/etiology , Postoperative Complications/immunology , Pressure , Rats , Rats, Inbred ACI , Rats, Inbred Strains , Transplantation, Heterotopic
12.
J Heart Lung Transplant ; 18(11): 1088-95, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10598732

ABSTRACT

INTRODUCTION: Reperfusion injury is an inflammatory cell-mediated response that causes tissue damage immediately following transplantation, and has been implicated in the development of acute and chronic rejection. NF-kappaB is a transcription factor that upregulates adhesion molecules ICAM-1, VCAM-1, and ELAM-1 following reperfusion. We hypothesized that treatment with sulfasalazine, a potent inhibitor of NF-kappaB, would decrease adhesion molecule expression, decrease reperfusion injury, and prolong allograft survival in rat cardiac transplants. METHODS: Heterotopic rat heart transplants were performed. Donor allografts were treated with saline, sulfasalazine (SSA), or lipopolysaccharide (LPS), a potent inducer of NF-kappaB activity. Reperfusion injury was assessed with cardiac edema (percent wet weight), neutrophil infiltration (MPO activity), and histologic damage (contraction band necrosis). Immunohistochemistry was performed to analyze protein expression. Acute rejection was determined by daily palpation. RESULTS: Treatment with a single 100 mg/kg intraperitoneal injection of sulfasalazine decreased reperfusion injury compared to saline controls (MPO activity, saline: 2.1+/-0.3, SSA: 1.2+/-0.31, P < 0.005; % wet weight, saline 77.6+/-1.1%; SSA 75.8+/-1.0%, P < 0.005; contraction band necrosis, saline: 13.1+/-2.5%, SSA: 6.1+/-3.4%, P < 0.001). LPS administration increased all parameters of reperfusion injury. Treatment with sulfasalazine prior to LPS also decreased reperfusion injury compared to LPS and saline groups. Sulfasalazine treatment decreased ICAM-1 and VCAM-1 protein expression. Administration of 500 mg/kg sulfasalazine increased graft survival to 15.4+/-1.8 days compared to saline (6.8+/-1.4 days, P < 0.005). CONCLUSION: Treatment with sulfasalazine is an effective method to decrease reperfusion injury and prolong allograft survival in a rat cardiac transplantation model.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Graft Rejection/prevention & control , Graft Survival/drug effects , Heart Transplantation , Myocardial Reperfusion Injury/prevention & control , NF-kappa B/antagonists & inhibitors , Sulfasalazine/therapeutic use , Animals , Disease Models, Animal , E-Selectin/biosynthesis , Graft Rejection/etiology , Graft Rejection/metabolism , Graft Rejection/pathology , Heart Transplantation/pathology , Injections, Intraperitoneal , Intercellular Adhesion Molecule-1/biosynthesis , Lipopolysaccharides/pharmacology , Male , Myocardial Reperfusion Injury/complications , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , NF-kappa B/metabolism , Rats , Rats, Inbred ACI , Transplantation, Homologous , Up-Regulation/drug effects , Vascular Cell Adhesion Molecule-1/biosynthesis
13.
J Heart Lung Transplant ; 11(2 Pt 1): 280-8, 1992.
Article in English | MEDLINE | ID: mdl-1576134

ABSTRACT

Correlation between the effective refractory period of the conduction system and the histopathologic grade of the ventricle was examined in the rat cardiac allograft during acute rejection. Lewis rats were recipients of intraabdominal heart grafts from brown Norway rats (allogeneic group, n = 42) or Lewis rats (syngeneic group, n = 15). No immunosuppressant was given. The effective refractory period of the conduction system was measured by the programmed atrial extrastimulus method (basic cycle length, 150 msec) just before the time of death. Specimens of the transplanted hearts were examined histopathologically and the histopathologic grade of rejection was scored according to the standardized grading system of the International Society for Heart and Lung Transplantation. The effective refractory period of the allogeneic heart was significantly longer starting on the third day after transplantation (p less than 0.01). The effective refractory period of the allogeneic heart was more prolonged as the severity of rejection increased. The correlation between the effective refractory period (Y) and the histopathologic grade (X) of the allogeneic group was statistically significant (r = 0.955; Y = 10.3X + 81.0; p less than 0.01). The effective refractory period of all syngeneic hearts and allogeneic hearts of postoperative day 1 and 2 were statistically equivalent to the period of native rat hearts (81.0 +/- 2.0 msec; n = 6). The histopathologic grade of the conduction system was the same as that of the ventricle. We conclude that the effective refractory period of the conduction system could be a useful measure to predict the histopathologic grade of cardiac allograft rejection.


Subject(s)
Cardiac Pacing, Artificial , Graft Rejection , Heart Conduction System/physiopathology , Heart Transplantation/immunology , Myocardium/pathology , Abdomen , Animals , Electrocardiography , Male , Rats , Rats, Inbred BN , Rats, Inbred Lew , Refractory Period, Electrophysiological/physiology , Transplantation, Heterotopic
14.
J Heart Lung Transplant ; 17(4): 349-55, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9588579

ABSTRACT

BACKGROUND: Chronic graft vascular disease in cardiac allografts results from coronary artery neointimal formation. Vascular ischemia has been shown to provoke the development of neointimal hyperplasia through endothelial damage. We used a rodent model of neointimal formation to test the in vivo effects of antisense oligodeoxynucleotides (AS ODN) specifically designed to block this process. METHODS: Aortas from ACI rats were mock transfected or transfected with 18 base pair AS ODNs against the 5' start codon region of both CDC2 kinase, and proliferating cell nuclear antigen (PCNA) mRNA. Transfection was accomplished by placing the aorta in ODN solution (transfected group, 40 micromol/L of each sequence) or saline solution alone (mock-transfected group) and exposing to hydrostatic pressure (2 atm) for 24 hours at 4 degrees C. Vessels were then interposition-grafted into the abdominal aorta of untreated isogenic recipients and procured on postoperative days (POD) 1, 6, and 14. RESULTS: Nuclear localization of fluorescein isothiocyanate ODN was observed in 81%+/-3% of medial smooth muscle cells at 24 hours after interposition grafting and reperfusion. Efficacy of AS ODNs at blocking CDC2 kinase and PCNA was verified on POD 6 by enzyme-linked immunosorbent assay. This blockade significantly reduced ischemically induced vascular narrowing on POD 14 as assessed by use of computerized image analysis (3.85%+/-2.45% luminal narrowing for transfected vs 7.11%+/-2.03% for control subjects, p < 0.03). CONCLUSIONS: Our data show the efficacy of AS ODN at blocking rat PCNA and CDC2 kinase up-regulation provoked by ischemia. This ex vivo approach had beneficial effects against vascular narrowing in a rodent model of ischemically induced neointimal hyperplasia, an antigen-independent factor important in the development of subsequent chronic graft vascular disease.


Subject(s)
Aorta, Abdominal/transplantation , Aortic Diseases/prevention & control , Oligonucleotides, Antisense/therapeutic use , Tunica Intima/pathology , Animals , CDC2 Protein Kinase/genetics , Chronic Disease , Codon/genetics , Coronary Disease/etiology , Endothelium, Vascular/pathology , Heart Transplantation/adverse effects , Hydrostatic Pressure , Hyperplasia , Image Processing, Computer-Assisted , Ischemia/physiopathology , Male , Proliferating Cell Nuclear Antigen/genetics , RNA, Messenger/genetics , Rats , Rats, Inbred ACI , Transfection , Transplantation, Isogeneic , Up-Regulation/drug effects
15.
Ann Thorac Surg ; 64(4): 1004-12, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9354518

ABSTRACT

BACKGROUND: The purpose of this study was to assess the impact of increased donor cardiac intercellular adhesion molecule (ICAM-1) expression on both reperfusion injury and chronic graft vascular disease after transplantation. METHODS: Hearts were harvested from donor rats before and after pretreatment with lipopolysaccharide at -24 hours, underwent 45 minutes of cold ischemia, and were transplanted into ACI recipients with or without anti-ICAM-1 monoclonal antibody treatment. Grafts were procured early for analysis of ICAM-1 expression and reperfusion injury or the recipients were treated with cyclosporin A (to allow long-term graft acceptance) for postoperative days 0 through 9 with procurement on postoperative day 90 to histologically score for chronic graft vascular disease. RESULTS: Lipopolysaccharide-pretreated PVG heart grafts showed increased ICAM-1 expression by Northern blot and immunohistochemical analysis leading to increased reperfusion injury as assessed by neutrophil infiltration (myeloperoxidase), cardiac edema (percentage wet weight), and histologic injury (percentage area of contraction band necrosis), which was reversed by recipient treatment with anti-ICAM-1 monoclonal antibody. After administration of cyclosporin A, 5 mg/kg for 10 days, lipopolysaccharide-treated grafts had significantly worse chronic graft vascular disease scores (2.56 +/- 0.57 versus 1.84 +/- 0.75; p < 0.05 by Mann-Whitney U test). CONCLUSIONS: The induction donor inflammatory state before harvest leading to increased cardiac ICAM-1 expression promotes reperfusion injury and chronic graft vascular disease after transplantation in this rodent heterotopic heart model.


Subject(s)
Coronary Disease/metabolism , Heart Transplantation/physiology , Intercellular Adhesion Molecule-1/metabolism , Myocardial Reperfusion Injury/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Chronic Disease , Coronary Disease/etiology , Cyclosporine/pharmacology , Disease Models, Animal , Heart Transplantation/adverse effects , Inflammation/chemically induced , Intercellular Adhesion Molecule-1/immunology , Lipopolysaccharides , Male , Myocardial Reperfusion Injury/etiology , Myocardium/metabolism , RNA, Messenger/genetics , Rats , Rats, Inbred ACI , Rats, Inbred Strains , Transplantation, Heterotopic , Up-Regulation
16.
Ann Thorac Surg ; 68(1): 119-24, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10421126

ABSTRACT

BACKGROUND: The goal of this study is to determine the effects of ex vivo hyperbaric pressure administration of AS-ICAM-1 ODN and systemic anti-LFA-1 mAb treatment on reperfusion injury in the rat cardiac allograft model. METHODS: A PVG to ACI functional heterotopic rat heart model was used. Donor hearts were treated with either saline or AS-ICAM-1 ODN and 5 atm of hyperbaric pressure for 45 minutes. Anti-LFA-1 mAb was administered systemically prior to reperfusion of the allograft. Allografts were procured 24 hours after transplantation for assessment of reperfusion injury or 72 hours to determine ICAM-1 protein expression. RESULTS: Ex vivo administration of AS-ICAM-1 ODN led to decreases in percentage wet weight (77.1+/-0.83% vs 78.7+/-1.0%, p < 0.05), myeloperoxidase activity (3.14+/-0.72 vs 4.07+/-0.59, p < 0.05), contraction band necrosis (6.4+/-6.47% vs 21.1+/-7.43%, p < 0.01), and ICAM-1 protein expression determined by immunohistochemistry compared to saline controls. Treatment with anti-LFA-1 mAb resulted in decreases in wet weight ratio (76.7+/-0.63%, p < 0.05 vs saline), myeloperoxidase activity (3.58+/-0.39, p < 0.05 vs saline) and contraction band necrosis (11.8+/-3.56%, p < 0.05 vs saline). Combination of pressure administration of AS-ICAM-1 ODN and anti-LFA-1 mAb decreased wet weight ratios (77.1+/-0.93%, p < 0.05 vs saline), myeloperoxidase activity (2.88+/-0.44, p < 0.01 vs saline), and contraction band necrosis (6.75+/-5.67%, p < 0.05 vs saline). CONCLUSIONS: Ex vivo pressure mediated delivery of AS-ICAM-1 ODN decreases ICAM-1 protein expression, reduces reperfusion injury in rodent cardiac allografts, and is more effective than anti-LFA-1 mAb treatment alone.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Heart Transplantation , Lymphocyte Function-Associated Antigen-1/immunology , Myocardial Reperfusion Injury/prevention & control , Oligodeoxyribonucleotides, Antisense/administration & dosage , Animals , Atmospheric Pressure , Intercellular Adhesion Molecule-1/analysis , Male , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Myocardium/pathology , Necrosis , Oligodeoxyribonucleotides , Peroxidase/metabolism , Rats , Rats, Inbred ACI , Rats, Inbred Strains , Transplantation, Heterotopic
20.
J Heart Transplant ; 7(5): 359-69, 1988.
Article in English | MEDLINE | ID: mdl-3058906

ABSTRACT

Cyclosporin A-associated nephrotoxicity has precipitated the need to develop new immunosuppressive protocols or agents that have a higher therapeutic index than cyclosporin A. A new immunosuppressive agent, cyclosporin G or norvaline (Nva-2) cyclosporine, has been shown to be potent. The rat heterotopic transplant model (ACI to Lewis) and Lewis rats that had no operation were used to compare cyclosporin G with cyclosporin A (5 mg/kg/day per gavage) with and without azathioprine (5 to 10 mg/kg/day, intraperitoneally) in terms of immunosuppressive efficacy (graft survival), toxicity (mortality, renal histopathology, and serum creatinine and blood urea nitrogen values), and pharmacokinetics (trough whole blood cyclosporine levels as measured by radioimmunoassay on days 14 and 28 of treatment). In this model no statistically significant difference in immunosuppression was shown between the two cyclosporins both with and without azathioprine. Cyclosporin G, however, was associated with significantly less mortality when combination therapy with azathioprine was used. Both cyclosporins were associated with normal serum creatinine values and little histopathologic evidence of nephrotoxicity, except juxtaglomerular apparatus hyperplasia. Comparable cyclosporine levels were achieved when cyclosporin A or G was used as the sole immunosuppressive agent, but significantly higher cyclosporine levels were shown with cyclosporin A than with cyclosporin G when combination therapy with azathioprine was used. Further studies in humans are needed to evaluate whether cyclosporin G will be a clinically useful immunosuppressive agent either alone or combined with other immunosuppressive modalities.


Subject(s)
Azathioprine/pharmacology , Cyclosporine , Cyclosporins/pharmacology , Immunosuppressive Agents/pharmacology , Animals , Azathioprine/toxicity , Cyclosporins/pharmacokinetics , Cyclosporins/toxicity , Drug Therapy, Combination , Heart Transplantation , Hyperplasia , Immunosuppressive Agents/pharmacokinetics , Immunosuppressive Agents/toxicity , Juxtaglomerular Apparatus/drug effects , Juxtaglomerular Apparatus/pathology , Male , Rats , Rats, Inbred Lew
SELECTION OF CITATIONS
SEARCH DETAIL