Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Int J Mol Sci ; 25(8)2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38674016

ABSTRACT

Organ transplantation is associated with various forms of programmed cell death which can accelerate transplant injury and rejection. Targeting cell death in donor organs may represent a novel strategy for preventing allograft injury. We have previously demonstrated that necroptosis plays a key role in promoting transplant injury. Recently, we have found that mitochondria function is linked to necroptosis. However, it remains unknown how necroptosis signaling pathways regulate mitochondrial function during necroptosis. In this study, we investigated the receptor-interacting protein kinase 3 (RIPK3) mediated mitochondrial dysfunction and necroptosis. We demonstrate that the calmodulin-dependent protein kinase (CaMK) family members CaMK1, 2, and 4 form a complex with RIPK3 in mouse cardiac endothelial cells, to promote trans-phosphorylation during necroptosis. CaMK1 and 4 directly activated the dynamin-related protein-1 (Drp1), while CaMK2 indirectly activated Drp1 via the phosphoglycerate mutase 5 (PGAM5). The inhibition of CaMKs restored mitochondrial function and effectively prevented endothelial cell death. CaMKs inhibition inhibited activation of CaMKs and Drp1, and cell death and heart tissue injury (n = 6/group, p < 0.01) in a murine model of cardiac transplantation. Importantly, the inhibition of CaMKs greatly prolonged heart graft survival (n = 8/group, p < 0.01). In conclusion, CaMK family members orchestrate cell death in two different pathways and may be potential therapeutic targets in preventing cell death and transplant injury.


Subject(s)
Dynamins , Graft Rejection , Heart Transplantation , Necroptosis , Receptor-Interacting Protein Serine-Threonine Kinases , Animals , Mice , Graft Rejection/metabolism , Graft Rejection/pathology , Heart Transplantation/adverse effects , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Dynamins/metabolism , Dynamins/genetics , Mitochondria/metabolism , Endothelial Cells/metabolism , Male , Mice, Inbred C57BL , Phosphoprotein Phosphatases/metabolism , Phosphoprotein Phosphatases/genetics , Phosphorylation , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Signal Transduction
2.
Int J Mol Sci ; 25(4)2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38396887

ABSTRACT

Kidney transplantation is preferred for end-stage renal disease. The current gold standard for kidney preservation is static cold storage (SCS) at 4 °C. However, SCS contributes to renal graft damage through ischemia-reperfusion injury (IRI). We previously reported renal graft protection after SCS with a hydrogen sulfide donor, sodium thiosulfate (STS), at 4 °C. Therefore, this study aims to investigate whether SCS at 10 °C with STS and Hemopure (blood substitute), will provide similar protection. Using in vitro model of IRI, we subjected rat renal proximal tubular epithelial cells to hypoxia-reoxygenation for 24 h at 10 °C with or without STS and measured cell viability. In vivo, we preserved 36 donor kidneys of Lewis rats for 24 h in a preservation solution at 10 °C supplemented with STS, Hemopure, or both followed by transplantation. Tissue damage and recipient graft function parameters, including serum creatinine, blood urea nitrogen, urine osmolality, and glomerular filtration rate (GFR), were evaluated. STS-treated proximal tubular epithelial cells exhibited enhanced viability at 10 °C compared with untreated control cells (p < 0.05). Also, STS and Hemopure improved renal graft function compared with control grafts (p < 0.05) in the early time period after the transplant, but long-term function did not reach significance. Overall, renal graft preservation at 10 °C with STS and Hemopure supplementation has the potential to enhance graft function and reduce kidney damage, suggesting a novel approach to reducing IRI and post-transplant complications.


Subject(s)
Hemoglobins , Kidney Transplantation , Reperfusion Injury , Thiosulfates , Rats , Animals , Organ Preservation , Graft Survival , Rats, Inbred Lew , Kidney , Reperfusion Injury/drug therapy , Reperfusion Injury/prevention & control
3.
Can J Surg ; 65(2): E193-E202, 2022.
Article in English | MEDLINE | ID: mdl-35292525

ABSTRACT

BACKGROUND: The increasing use of kidneys from donations after cardiac death (DCD) for renal transplantation is hindered by negative outcomes owing to organ injury after prolonged warm and cold ischemia-reperfusion. Recently, hydrogen sulfide (H2S) has shown cytoprotective effects against ischemia-reperfusion injury; however, its effectiveness in the context of DCD renal transplantation is unknown. METHODS: We tested a novel 30-day in vivo syngeneic murine model of DCD renal transplantation, in which the donor kidney was clamped for 30 minutes and stored for 18 hours in cold University of Wisconsin (UW) solution or UW with 150 µM sodium hydrogen sulfide (UW + NaHS) before transplantation. We also tested a 7-day in vivo porcine model of DCD renal autotransplantation, in which the left kidney was clamped for 60 minutes and preserved for 24 hours using hypothermic perfusion with UW or UW + 150 µM NaHS before autotransplantation. We collected blood and urine samples periodically, and collected kidney samples at the end point for histopathology and quantitative reverse transcription polymerase chain reaction. RESULTS: Rats that received H2S-treated kidneys showed significantly higher survival, faster recovery of graft function and significantly lower acute tubular necrosis than controls. Pig kidneys perfused with UW + NaHS showed significantly higher renal blood flow and lower renal resistance than control kidneys after 24 hours of perfusion. After autotransplantation, pigs that received H2S-treated kidneys showed significantly lower serum creatinine on days 1 and 7 after transplantation. Rat and pig kidneys treated with H2S also showed more protective gene expression profiles than controls. CONCLUSION: Our findings support the potential use of H2S-supplemented UW solution during cold storage as a novel and practical means to improve DCD graft survival and function.


Subject(s)
Hydrogen Sulfide , Kidney Transplantation , Organ Preservation Solutions , Reperfusion Injury , Adenosine , Allopurinol , Animals , Death , Glutathione , Humans , Hydrogen Sulfide/pharmacology , Insulin , Kidney/blood supply , Mice , Organ Preservation Solutions/pharmacology , Raffinose , Rats , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Swine
4.
Am J Transplant ; 21(10): 3268-3279, 2021 10.
Article in English | MEDLINE | ID: mdl-33784431

ABSTRACT

Inflammation posttransplant is directly linked to cell death programs including apoptosis and necrosis. Cell death leads to the release of cellular contents which can promote inflammation. Targeting of these pathways should be an effective strategy to prevent transplant rejection. Toll-like receptor 3 (TLR3) is emerging as a major endogenous sensor of inflammation. In this study, we assessed the role of TLR3 on cell death and transplant rejection. We showed that TLR3 is highly expressed on mouse microvascular endothelial cell (ECs) and the endothelium of cardiac grafts. We demonstrated that TLR3 interacting with dsRNA or self-RNA triggered apoptosis and necroptosis in ECs. Interestingly, TLR3-induced necroptosis led mitochondrial damage. Inhibition of the mitochondrial membrane permeability molecule Cyclophilin D prevented necroptosis in ECs. In vivo, endothelium damage and activities of caspase-3 and mixed lineage kinase domain-like protein were inhibited in TLR3-/- cardiac grafts compared with C57BL/6 grafts posttransplant (n = 5, p < .001). Importantly, TLR3-/- cardiac grafts had prolonged survival in allogeneic BALB/c mice (mean survival = 121 ± 67 vs. 31 ± 6 days of C57BL/6 grafts, n = 7, p = .002). In summary, our study suggests that TLR3 is an important cell death inducer in ECs and cardiac grafts and thus a potential therapeutic target in preventing cardiac transplant rejection.


Subject(s)
Heart Transplantation , Toll-Like Receptor 3 , Animals , Apoptosis , Cell Death , Heart Transplantation/adverse effects , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tissue Donors , Toll-Like Receptor 3/metabolism
5.
Int J Mol Sci ; 22(20)2021 Oct 13.
Article in English | MEDLINE | ID: mdl-34681708

ABSTRACT

Ischemia-reperfusion injury (IRI) is an inevitable consequence of organ transplant procedure and associated with acute and chronic organ rejection in transplantation. IRI leads to various forms of programmed cell death, which worsens tissue damage and accelerates transplant rejection. We recently demonstrated that necroptosis participates in murine cardiac microvascular endothelial cell (MVEC) death and murine cardiac transplant rejection. However, MVEC death under a more complex IRI model has not been studied. In this study, we found that simulating IRI conditions in vitro by hypoxia, reoxygenation and treatment with inflammatory cytokines induced necroptosis in MVECs. Interestingly, the apoptosis-inducing factor (AIF) translocated to the nucleus during MVEC necroptosis, which is regulated by the mitochondrial permeability molecule cyclophilin D (CypD). Furthermore, CypD deficiency in donor cardiac grafts inhibited AIF translocation and mitigated graft IRI and rejection (n = 7; p = 0.002). Our studies indicate that CypD and AIF play significant roles in MVEC necroptosis and cardiac transplant rejection following IRI. Targeting CypD and its downstream AIF may be a plausible approach to inhibit IRI-caused cardiac damage and improve transplant survival.


Subject(s)
Apoptosis Inducing Factor/metabolism , Necroptosis , Peptidyl-Prolyl Isomerase F/metabolism , Animals , Apoptosis Inducing Factor/antagonists & inhibitors , Apoptosis Inducing Factor/genetics , Cell Hypoxia , Cell Nucleus/metabolism , Peptidyl-Prolyl Isomerase F/deficiency , Peptidyl-Prolyl Isomerase F/genetics , Endothelial Cells/cytology , Endothelial Cells/metabolism , Interferon-gamma/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microvessels/cytology , Models, Biological , Necroptosis/drug effects , Oxygen/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Tumor Necrosis Factor-alpha/pharmacology
6.
Am J Transplant ; 19(3): 686-698, 2019 03.
Article in English | MEDLINE | ID: mdl-30203531

ABSTRACT

Transplantation is invariably associated with programmed cell death including apoptosis and necrosis, resulting in delayed graft function and organ rejection. We have demonstrated the contribution of necroptosis to mouse microvascular endothelial cell (MVEC) death and transplant rejection. Organ injury results in the opening of mitochondrial permeability transition pores (mPTPs), which can trigger apoptotic molecules release that ultimately results in cell death. The effect of mPTPs in the necroptotic pathway remains controversial; importantly, their role in transplant rejection is not clear. In this study, tumor necrosis factor-α triggered MVECs to undergo receptor-interacting protein kinase family (RIPK1/3)-dependent necroptosis. Interestingly, inhibition of mPTP opening could also inhibit necroptotic cell death. Cyclophilin-D (Cyp-D) is a key regulator of the mPTPs. Both inhibition and deficiency of Cyp-D protected MVECs from necroptosis (n = 3, P < .00001). Additionally, inhibition of Cyp-D attenuated RIPK3-downstream mixed-lineage kinase domain-like protein phosphorylation. In vivo, Cyp-D-deficient cardiac grafts showed prolonged survival in allogeneic BALB/c mice posttransplant compared with wild-type grafts (n = 7, P < .0001). Our study results suggest that the mPTPs may be important mechanistic mediators of necroptosis in cardiac grafts. There is therapeutic potential in targeting cell death via inhibition of the mPTP-regulating molecule Cyp-D to prevent cardiac graft rejection.


Subject(s)
Cell Membrane Permeability , Endothelial Cells/pathology , Graft Rejection/etiology , Heart Transplantation/adverse effects , Mitochondria/pathology , Necroptosis , Peptidyl-Prolyl Isomerase F/metabolism , Allografts , Animals , Peptidyl-Prolyl Isomerase F/genetics , Endothelial Cells/immunology , Endothelial Cells/metabolism , Graft Rejection/metabolism , Graft Rejection/pathology , Graft Survival , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mitochondria/immunology , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tissue Donors , Tumor Necrosis Factor-alpha/pharmacology
7.
Am J Transplant ; 19(11): 3139-3148, 2019 11.
Article in English | MEDLINE | ID: mdl-31338943

ABSTRACT

Heart transplant has been accepted as the standard treatment for end-stage heart failure. Because of its susceptibility to ischemia-reperfusion injury, the heart can be preserved for only 4 to 6 hours in cold static preservation solutions. Prolonged ischemia time is adversely associated with primary graft function and long-term survival. New strategies to preserve donor hearts are urgently needed. We demonstrate that AP39, a mitochondria-targeting hydrogen sulfide donor, significantly increases cardiomyocyte viability and reduces cell apoptosis/death after cold hypoxia/reoxygenation in vitro. It also decreases gene expression of proinflammatory cytokines and preserves mitochondria function. Using an in vivo murine heart transplant model, we show that preserving donor hearts with AP39-supplemented University of Wisconsin solution (n = 7) significantly protects heart graft function, measured by quantitative ultrasound scan, against prolonged cold ischemia-reperfusion injury (24 hours at 4°C), along with reducing tissue injury and fibrosis. Our study demonstrates that supplementing preservation solution with AP39 protects cardiac grafts from prolonged ischemia, highlighting its therapeutic potential in preventing ischemia-reperfusion injury in heart transplant.


Subject(s)
Heart Transplantation/methods , Hydrogen Sulfide/metabolism , Mitochondria/drug effects , Organ Preservation Solutions/administration & dosage , Organ Preservation/methods , Organophosphorus Compounds/pharmacology , Reperfusion Injury/prevention & control , Thiones/pharmacology , Animals , Male , Mice , Mice, Inbred C57BL , Mitochondria/pathology , Tissue Donors/supply & distribution
8.
Nephrology (Carlton) ; 24(6): 661-669, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30175514

ABSTRACT

BACKGROUND: Ischaemia-reperfusion injury (IRI) is associated with programmed cell death that promotes inflammation and organ dysfunction. Necroptosis is mediated by members of receptor interacting protein kinases (RIPK1/3). Inhibition of RIPK1/3 provides a pro-survival benefit in kidney IRI. Caspase-8 initiates apoptosis and contributes to IRI. We studied whether inhibiting both RIPK3 and caspase-8 would provide an additional benefit in kidney IRI. METHODS: A clamp was applied to the left kidney pedicle for 45 min followed by right kidney nephrectomy. Kidney and serum from wild type, RIPK3-/- , and RIPK3-/- caspase-8-/- double knockout (DKO) mice were collected post-IRI for assessment of injury. Tubular epithelial cells (TEC) isolated from wild type, RIPK3-/- , and DKO mice were treated with interferons-γ and interleukin-1ß to induce apoptotic death. RESULTS: Kidney IRI of DKO mice did not show improvement over RIPK3-/- mice. We have found that DKO triggered 'intrinsic' apoptosis in TEC in response to interleukin-1ß and interferons-γ. Up-regulation of the B-cell lymphoma 2 (Bcl-2)-associated death promoter, the Bcl-2-homologous antagonist killer and Bcl-2-associated X protein and enhanced activation of caspase-3 and 9 were found in DKO TEC. TEC infected with Murine cytomegalovirus that encodes multiple cell death inhibitors resist to death. CONCLUSION: We show that the deletion of both RIPK3 and caspase-8 does not provide additive benefit in IRI or TEC death and may enhance injury by up-regulation of intrinsic apoptosis. This suggests blocking multiple death pathways may be required for the prevention of kidney IRI clinically.


Subject(s)
Apoptosis , Caspase 8/metabolism , Epithelial Cells/enzymology , Kidney Diseases/enzymology , Kidney Tubules/enzymology , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Reperfusion Injury/enzymology , Animals , Apoptosis/drug effects , Caspase 8/genetics , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/pathology , Interferon-gamma/pharmacology , Interleukin-1beta/pharmacology , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney Tubules/drug effects , Kidney Tubules/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Necroptosis , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Signal Transduction
9.
J Urol ; 196(6): 1778-1787, 2016 12.
Article in English | MEDLINE | ID: mdl-27177428

ABSTRACT

PURPOSE: Chronic obstructive uropathy can cause irreversible kidney injury, atrophy and inflammation, which can ultimately lead to fibrosis. Epithelial-mesenchymal transition is a key trigger of fibrosis that is caused by up-regulation of TGF-ß1 (transforming growth factor-ß1) and ANGII (angiotensin II). H2S is an endogenously produced gasotransmitter with cytoprotective properties. We sought to elucidate the effects of the slow-releasing H2S donor GYY4137 on chronic ureteral obstruction and evaluate the potential mechanisms. MATERIALS AND METHODS: Following unilateral ureteral obstruction male Lewis rats were given daily intraperitoneal administration of phosphate buffered saline vehicle (obstruction group) or phosphate buffered saline plus 200 µmol/kg GYY4137 (obstruction plus GYY4137 group) for 30 days. Urine and serum samples were collected to determine physiological parameters of renal function and injury. Kidneys were removed on postoperative day 30 to evaluate histopathology and protein expression. Epithelial-mesenchymal transition in LLC-PK1 pig kidney epithelial cells was induced with TGF-ß1 and treated with GYY4137 to evaluate potential mechanisms via in vitro scratch wound assays. RESULTS: H2S treatment decreased serum creatinine and the urine protein-to-creatinine excretion ratio after unilateral ureteral obstruction. In addition, H2S mitigated cortical loss, inflammatory damage and tubulointerstitial fibrosis. Tissues showed decreased expression of epithelial-mesenchymal transition markers upon H2S treatment. Epithelial-mesenchymal transition progression in LLC-PK1 was alleviated upon in vitro administration of GYY4137. CONCLUSIONS: To our knowledge our findings demonstrate for the first time the protective effects of H2S in chronic obstructive uropathy. This may represent a potential therapeutic solution to ameliorate renal damage and improve the clinical outcomes of urinary obstruction.


Subject(s)
Hydrogen Sulfide/therapeutic use , Kidney Diseases/etiology , Kidney Diseases/prevention & control , Morpholines/therapeutic use , Organothiophosphorus Compounds/therapeutic use , Ureteral Obstruction/complications , Animals , Chronic Disease , Male , Rats , Rats, Inbred Lew , Swine
10.
J Am Soc Nephrol ; 22(1): 82-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21051739

ABSTRACT

Disruption of the dopamine D(5) receptor gene in mice increases BP and causes salt sensitivity. To determine the role of renal versus extrarenal D(5) receptors in BP regulation, we performed cross-renal transplantation experiments. BP was similar between wild-type mice and wild-type mice transplanted with wild-type kidneys, indicating that the transplantation procedure did not affect BP. BP was lower among D(5)(-/-) mice transplanted with wild-type kidneys than D(5)(-/-) kidneys, demonstrating that the renal D(5) receptors are important in BP control. BP was higher in wild-type mice transplanted with D(5)(-/-) kidneys than wild-type kidneys but not significantly different from syngenic transplanted D(5)(-/-) mice, indicating the importance of the kidney in the development of hypertension. On a high-salt diet, all mice with D(5)(-/-) kidneys excreted less sodium than mice with wild-type kidneys. Transplantation of a wild-type kidney into a D(5)(-/-) mouse decreased the renal expression of AT(1) receptors and Nox-2. Conversely, transplantation of a D(5)(-/-) kidney into a wild-type mouse increased the expression of both, suggesting that both renal and extrarenal factors are important in the regulation of AT(1) receptor and Nox-2 expression. These results highlight the role of renal D(5) receptors in BP homeostasis and the pathogenesis of hypertension.


Subject(s)
Blood Pressure/physiology , Hypertension/etiology , Hypertension/metabolism , Kidney/metabolism , Receptors, Dopamine D5/deficiency , Animals , Blood Pressure/drug effects , Disease Models, Animal , Hypertension/physiopathology , Kidney/drug effects , Kidney Transplantation , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , NADPH Oxidase 2 , NADPH Oxidases/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptors, Dopamine D5/genetics , Receptors, Dopamine D5/metabolism , Sodium/urine , Sodium Chloride, Dietary/pharmacology
11.
J Immunol ; 182(10): 5970-81, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19414748

ABSTRACT

We report on a novel approach aimed at preventing acute vascular rejection (AVR), one of the major unresolved hurdles of clinical transplantation. In a C3H-to-BALB/c heterotopic heart transplant model, we demonstrate that free bone transplantation combined with cyclosporin A suppresses antidonor Ab responses, induces indefinite cardiac allograft survival (>100 days), and preserves graft architecture. In contrast, untreated- or cyclosporin A alone-treated recipients rejected their cardiac grafts on days 7.7 +/- 0.6 and 15.5 +/- 1.1, respectively, with graft histology indicative of AVR. Splenic dendritic cells from nonrejecting recipients expressed low levels of MHC II, CD40, and CD86, reduced ability to stimulate donor cell proliferation, and augmented IL-10 production of responding T cells in vitro. Adoptive transfer of dendritic cells from long-term surviving recipients 1 day before cardiac grafting was able to confer hyporesponsiveness to naive BALB/c recipients of cardiac allografts. To determine whether graft survival was associated with hematopoietic or stromal elements of the transplanted free bone, we administered isolated bone marrow mononuclear cells or free bone that was irradiated to deplete hematopoietic elements. Although bone marrow mononuclear cells had no effect on cardiac graft survival, irradiated free bone transplantation was capable of prolonging graft survival. Most interestingly, the prolongation effect was Ag nonspecific, because third party irradiated bone graft was also effective. Due to the fact that current immunosuppressive approaches are clinically ineffective at preventing AVR, this study provides promise for further investigations of BM components as a means of addressing a currently unmet medical need.


Subject(s)
Bone Transplantation/immunology , Cyclosporine/therapeutic use , Graft Rejection/prevention & control , Graft Survival/immunology , Heart Transplantation , Immunosuppressive Agents/therapeutic use , Adoptive Transfer , Animals , Bone Marrow Transplantation/immunology , Cytokines/biosynthesis , Cytokines/immunology , Dendritic Cells/immunology , Dendritic Cells/transplantation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Graft Rejection/immunology , Graft Survival/drug effects , Immunohistochemistry , Lymphocyte Culture Test, Mixed , Male , Mice , Transplantation, Homologous
12.
Transplantation ; 105(2): 308-317, 2021 02 01.
Article in English | MEDLINE | ID: mdl-32776778

ABSTRACT

BACKGROUND: Ischemia-reperfusion injury (IRI) is the major cause of primary graft dysfunction in organ transplantation. The mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) signaling pathway plays a crucial role in cell physiological and pathological processes including IRI. This study aims to investigate whether inhibition of ERK signaling with U0126 can prevent prolonged cold IRI in heart transplantation. METHODS: Rat cardiac cell line H9c2 cells were treated with U0126 before exposure to hypothermic hypoxia/reoxygenation (H/R) conditions. The effect of U0126 on H9c2 cells in response to H/R stress was determined by measuring cell death, reactive oxygen species production, mitochondrial membrane potential, and ERK signaling activation. Mouse syngeneic heterotopic heart transplantation was conducted, where a donor heart was preserved in the University of Wisconsin (UW) solution supplemented with U0126 for 24 hours at 4°C before transplantation. Heart graft function, histopathologic changes, apoptosis, and fibrosis were measured to assess IRI. RESULTS: Phosphorylated ERK was increased in both in vitro H/R-injured H9c2 cells and in vivo heart grafts with IRI. Pretreatment with U0126 inhibited ERK phosphorylation and prevented H9c2 cells from cell death, reactive oxygen species generation, and mitochondrial membrane potential loss in response to H/R. Preservation of donor hearts with U0126-supplemented solution improved graft function and reduced IRI by reductions in cell apoptosis/death, neutrophil infiltration, and fibrosis of the graft. CONCLUSIONS: Addition of U0126 to UW solution reduces ERK signal activation and attenuates prolonged cold IRI in a heart transplantation model. ERK inhibition with U0126 may be a useful strategy to minimize IRI in organ transplantation.


Subject(s)
Butadienes/pharmacology , Cold Ischemia , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Heart Transplantation/adverse effects , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Nitriles/pharmacology , Organ Preservation Solutions/pharmacology , Organ Preservation , Protein Kinase Inhibitors/pharmacology , Adenosine/pharmacology , Allopurinol/pharmacology , Animals , Apoptosis/drug effects , Cell Hypoxia , Cell Line , Cold Ischemia/adverse effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibrosis , Glutathione/pharmacology , Insulin/pharmacology , Male , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred C57BL , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Organ Preservation/adverse effects , Oxidative Stress/drug effects , Phosphorylation , Raffinose/pharmacology , Rats , Signal Transduction , Ventricular Function, Left/drug effects
13.
Transpl Immunol ; 19(2): 93-102, 2008 May.
Article in English | MEDLINE | ID: mdl-18503884

ABSTRACT

BACKGROUND: Regulatory T (Treg) cells are the immune suppressors in the maintenance of immune homeostasis and tolerance to self and non-self antigens, and may have therapeutic potential in the treatment of transplant rejection in patients. However, Treg cell development and action are poorly understood in transplantation. In this study, the association of CD4(+)Foxp3(+) infiltrates within renal allograft tissue with graft survival was investigated in a mouse model. METHODS: Kidney donors from C57BL/6J mice (H-2(b)) were transplanted to bilaterally nephrectomized Balb/c recipient mice (H-2(d)). Treg cells were examined with FACS and immunohistochemical staining. RESULTS: Here we showed that without any immunosuppressive regimen, kidney allografts were mostly rejected from 20 to 60 days after transplantation. During the progression of allograft rejection Foxp3(+) Treg phenotype infiltrates were significantly diminished, while intragraft expression of TGF-beta1, IL-6, IL-17 and IL-23 was up-regulated. The regulatory function of CD4(+)CD25(+) infiltrates was confirmed by their suppressive activity in mixed lymphocyte reaction. Further in vitro studies indicated that primary renal tubular epithelial cell (TEC) cultures produced high levels of IL-6 in response to allogeneic lymphocyte or IL-17 stimulation, and neutralization of IL-6 increased CD4(+)CD25(+)Foxp3(+) cells in co-cultures with TEC. CONCLUSION: Diminution of Foxp3(+) Treg infiltrates associates with renal allograft rejection, and neutralization of IL-6 activity enhances Foxp3(+) Treg cell differentiation. Our findings suggest that increase in intragraft IL-6 may down-regulate infiltrating Foxp3(+) Treg cells.


Subject(s)
Forkhead Transcription Factors/metabolism , Graft Rejection/immunology , Kidney Transplantation/immunology , T-Lymphocytes, Regulatory/immunology , Transplantation Tolerance/immunology , Animals , Disease Models, Animal , Disease Progression , Forkhead Transcription Factors/immunology , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukin-23/immunology , Interleukin-23/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Kidney/immunology , Lymphocyte Activation , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/metabolism , Transplantation, Homologous
14.
Microsurgery ; 28(3): 197-202, 2008.
Article in English | MEDLINE | ID: mdl-18286660

ABSTRACT

We have developed a mouse duodenojejunal bypass (DJB) surgical model that is for studying the effects of bariatric surgery on glucose homeostasis and has potential to impact clinical therapy of diabetes. The operation consists of using the majority of the duodenum and proximal part of the jejunum for biliopancreatic diversion. The distal end of the jejunum is anastomosed in an end-to-end fashion to the remaining proximal end of the duodenum just distal to the pylorus. The biliopancreatic secretions are diverted into the distal jejunum through an end-to-side anastomosis. We performed 10 DJB operations in C57BL/6 mice, with a 100% survival rate. The surgery had no effect on the growth or feeding patterns of the animals. The intestinal mucosa showed normal histology and function. This study confirms that it is technically possible to perform DJB surgery in mice. This mouse model can be used in the study of surgical treatment for type II diabetes.


Subject(s)
Bariatric Surgery/methods , Biliopancreatic Diversion/methods , Diabetes Mellitus/surgery , Duodenum/surgery , Jejunum/surgery , Anastomosis, Surgical/methods , Animals , Bariatric Surgery/adverse effects , Blood Glucose/physiology , Diabetes Mellitus/metabolism , Homeostasis , Male , Mice , Mice, Inbred C57BL , Models, Animal , Random Allocation
15.
Transplantation ; 84(9): 1158-67, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17998872

ABSTRACT

BACKGROUND: Innate immunity provides obstacles to successful organ transplantation, which cannot be prevented by cyclosporine (CsA). Here we have determined the potential of a myxoma viral serpin, Serp-1, with proven anti-inflammatory and antiatherogenic actions, to modulate innate immunity and contribute synergistically with CsA in the prevention of acute cardiac allograft rejection. METHODS: Brown-Norway rat hearts were heterotopically transplanted into Lewis rats and given either a monotherapy treatment of Serp-1, a subtherapeutic dose of CsA, or the two drugs in combination. RESULTS: A brief treatment of Serp-1 alone, or a subtherapeutic dose of CsA, resulted in a marked decrease in intragraft macrophage infiltration and downregulation of toll-like receptor (TLR)-2, TLR4 and MyD88 at 48 hours posttransplantation, which was associated with significantly reduced numbers of mature dendritic cells. A significant reduction in intragraft T-lymphocyte infiltration was observed with both Serp-1 monotherapy and Serp-1 and CsA combination therapy, with the combination treatment achieving indefinite graft survival (>100 days) with normal histology. The CsA monotherapy group displayed partially reduced lymphocyte infiltration compared to the untreated controls, but failed to inhibit early innate immune graft recognition events such as macrophage infiltration and TLR 2, TLR4, and MyD88, and was ultimately unsuccessful in preventing rejection (36.3+/-7.8 days). CONCLUSION: Observed suppressive effects of Serp-1 on early innate immune response components such as TLR-2 and 4, and on adaptive responses such as T-cell intragraft infiltration suggests that Serp-1 may modulate the transition from innate to adaptive immunity, exhibiting a synergistic effect on allograft survival when used in combination with a subtherapeutic dose of CsA.


Subject(s)
Graft Survival/physiology , Heart Transplantation/physiology , Serpins/therapeutic use , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Animals , Cyclosporine/therapeutic use , Down-Regulation , Heart Transplantation/immunology , Heart Transplantation/pathology , Immunosuppressive Agents , Male , Models, Animal , Rats , Rats, Inbred BN , Toll-Like Receptor 2/drug effects , Toll-Like Receptor 4/drug effects , Transplantation, Homologous
16.
Transplantation ; 84(12): 1636-43, 2007 Dec 27.
Article in English | MEDLINE | ID: mdl-18165776

ABSTRACT

BACKGROUND: The purpose of this study was to determine if a short course of monoclonal antibody (mAb) against CD45RB, LF 15-0195, and rapamycin would achieve long-term survival by inducing tolerance in a mouse limb transplant model. METHODS: Group 1 (n=9) consisted of nine isogenic (C57BL/6) transplants. Group 2 (n=3) included C57BL/6-to-BALB/c transplants receiving no drug therapy. Group 3 mice (n=4) were treated with mAb (3 mg/kg) and LF (2 mg/kg), and Group 4 (n=13) was treated with mAb, LF, and rapamycin (2 mg/kg). Both treatment groups received drug treatment for only 14 days posttransplantation. Animals were sacrificed if they displayed evidence of rejection or when deemed to be tolerant (defined as >day 100). RESULTS: All isografts had normal histology and graft function on day 100. Untreated C57BL/6-to-BALB/c allografts developed acute rejection within 10 days. The combination of mAb and LF prolonged allograft survival to a mean of 39+/-7 days. In Group 4, two animals had to be sacrificed at days 28 and 76 due to acute urinary retention. Transplant tolerance was achieved in 8 of the remaining 11 animals with a mean survival time of 100+/-12 days. Donor specific tolerance was demonstrated through permanent acceptance of skin grafts from the donor strain and rejection of skin grafts from C3H mice. Three Group 4 animals showed clinical and histological signs of mild, chronic rejection. Dendritic cells isolated from tolerant recipients exerted a suppressive effect in mixed lymphocyte reaction. CONCLUSION: A short course of anti-CD45RB mAb and LF 15-0195 prolonged limb allograft survival. The addition of rapamycin induced limb allograft tolerance which is associated with the generation of tolerogenic dendritic cells that suppressed T-cell proliferation.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Extremities/transplantation , Graft Survival/physiology , Guanidines/therapeutic use , Immunosuppressive Agents/therapeutic use , Leukocyte Common Antigens/immunology , Sirolimus/therapeutic use , Skin Transplantation/immunology , Transplantation, Homologous/immunology , Transplantation, Homologous/pathology , Animals , Isoantibodies/blood , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Animal , Transplantation Chimera
17.
Transplantation ; 101(9): 2026-2037, 2017 09.
Article in English | MEDLINE | ID: mdl-29633982

ABSTRACT

BACKGROUND: Despite advances in immunosuppressive therapies, the rate of chronic transplant loss remains substantial. Organ injury involves various forms of cell death including apoptosis and necrosis. We now recognize that early injury of cardiac transplants involves a newly described form of programmed necrotic cell death, termed necroptosis. Because this involves receptor-interacting protein (RIP) kinase 1/3, this study aimed to establish the role of RIP3 in chronic cardiac allograft rejection. METHODS: We used major histocompatibility complex class II mismatched C57BL/6N (H-2; B6) or B6.RIP3 (H-2; RIP3) mice to B6.C-H-2 (H2-Ab1; bm12) mouse cardiac transplantation. Microvascular endothelial cells (MVEC) were developed from B6 and RIP3 cardiac grafts. RESULT: CD4 T cell-mediated cardiac graft rejection is inhibited using RIP3 deficient donor grafts, with reduced cellular infiltration and vasculopathy compared with wild type cardiac grafts. Alloreactive CD4 T cell-mediated MVEC death involves TNFα, Fas ligand (FasL) and granzyme B. Although necroptosis and release of danger molecule high-mobility group box 1 are eliminated by the absence of RIP3, CD4 T cells had attenuated MVEC death through granzyme B and FasL. CONCLUSIONS: CD4 T cell-mediated MVEC death involves in TNFα, FasL and granzyme B. Necroptotic cell death and release of the danger molecule may promote inflammatory responses and transplant rejection. Although loss of RIP3 does not eliminate alloimmune responses, chronic graft injury is reduced. RIP3 is an important therapeutic target but additional granzyme and caspases inhibition is required for sufficiently improving long-term graft survival.


Subject(s)
Apoptosis , CD4-Positive T-Lymphocytes/metabolism , Cytotoxicity, Immunologic , Endothelial Cells/enzymology , Graft Rejection/enzymology , Heart Transplantation/adverse effects , Microvessels/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Allografts , Animals , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Endothelial Cells/immunology , Endothelial Cells/pathology , Fas Ligand Protein/metabolism , Graft Rejection/immunology , Graft Rejection/pathology , Graft Rejection/prevention & control , Granzymes/metabolism , Lymphocyte Activation , Male , Mice, Inbred C57BL , Mice, Knockout , Microvessels/metabolism , Microvessels/pathology , Necrosis , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction , Time Factors , Tumor Necrosis Factor-alpha/metabolism
18.
Oncotarget ; 8(22): 36531-36544, 2017 May 30.
Article in English | MEDLINE | ID: mdl-28388574

ABSTRACT

Ischemia reperfusion (I/R) injury which inevitably occurs during heart transplantation is the major factor leading to organ failure and graft rejection. In order to develop new therapies to prevent I/R injury, we used both a murine heart transplantation model with 24 hour cold I/R and an in vitro cell culture system to determine whether growth differentiation factor 15 (GDF15) is a protective factor in preventing I/R injury in heart transplantation and to further investigate underlying mechanisms of I/R injury. We found that cold I/R caused severe damage to the endocardium, epicardium and myocardium of heart grafts from wild type C57BL/6 mice, whereas grafts from GDF15 transgenic (TG) mice showed less damage as demonstrated by decreased cell apoptosis/death, decreased neutrophils infiltration and the preservation of the normal structure of the heart. Over-expression of GDF15 reduced expression of phosphorylated RelA p65, pre-inflammatory and pro-apoptotic genes while it enhanced Foxo3a phosphorylation in vitro and in vivo. Over-expression of GDF15 inhibited cell apoptosis/death and reduced neutrophil infiltration. In conclusion, this study, for the first time, demonstrates that GDF15 is a promising target for preventing cold I/R injury in heart transplantation. This study also shows that the resultant protective effects are mediated by the Foxo3 and NFκB signaling pathways.


Subject(s)
Cold Ischemia/adverse effects , Forkhead Box Protein O3/metabolism , Gene Expression , Growth Differentiation Factor 15/genetics , Heart Transplantation , Reperfusion Injury/etiology , Reperfusion Injury/metabolism , Animals , Apoptosis/genetics , Cytokines/genetics , Cytokines/metabolism , Humans , Inflammation Mediators/metabolism , Male , Mice , Myocytes, Cardiac/metabolism , Peroxidase/metabolism , Phosphorylation , Rats , Reperfusion Injury/prevention & control , Signal Transduction
19.
Transplantation ; 82(3): 406-14, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16906041

ABSTRACT

BACKGROUND: Chronic allograft nephropathy is a sclerotic process characterized by an increased extracellular matrix (ECM) protein deposition. Fibronectin (FN) is a major component of ECM. FN has been reported to undergo alternative splicing and produce several isoforms including the extra domain-B (ED-B) containing embryonic isoform. In the present study, we investigated ED-B FN expression in chronic allograft nephropathy and its relationship with endothelins (ET). METHODS: To establish chronic allograft nephropathy, allografts were performed between Fisher 344 --> Lewis rats. Allograft recipients were then randomly divided into two groups, allografts and allografts treated with ET receptor antagonist bosentan. Lewis --> Lewis recipients were used as isograft controls. Grafts were harvested at 30, 90 and 140 days for histological and gene expression analyses with respect to ED-B FN, ET-1 and transforming growth factor-beta1 (TGF-beta1) mRNA. ED-B FN protein levels were assessed by immunohistochemical analysis. Additionally, we analyzed human renal allograft biopsies. RESULTS: Our data demonstrates that rat chronic allograft nephropathy is associated with progressive upregulation of ED-B FN mRNA and protein. ET-1 and TGF-beta1 mRNA were also upregulated. Treatment of allograft recipient rats with bosentan prevented upregulation of ED-B FN and TGF-beta1. We further show that total FN, ED-B FN, ET-1 and TGF-beta1 mRNA expression were upregulated in human chronic allograft nephropathy specimens. CONCLUSION: Results obtained from both human and rat renal allograft tissues suggest that expression of ED-B FN is upregulated in chronic allograft nephropathy and such upregulation is mediated via ET-1 and its interaction with TGF-beta1.


Subject(s)
Endothelin-1/metabolism , Fibronectins/metabolism , Kidney Transplantation , Nephritis/metabolism , Nephritis/pathology , Adolescent , Adult , Aged , Animals , Biopsy , Child , Chronic Disease , Endothelin-1/genetics , Female , Fibronectins/genetics , Gene Expression Regulation , Graft Rejection , Humans , Kidney/metabolism , Kidney/physiopathology , Male , Middle Aged , Nephritis/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , Rats , Transforming Growth Factor beta/genetics , Transplantation, Homologous
20.
Transplantation ; 81(2): 265-72, 2006 Jan 27.
Article in English | MEDLINE | ID: mdl-16436971

ABSTRACT

BACKGROUND: Xenotransplantation may provide a solution to the increasing shortage of donor organs. Acute vascular rejection and cell-mediated rejection remain the primary barriers to successful xenotransplantation. In animal models where acute vascular rejection can be attenuated, xenografts succumb to cell-mediated rejection. The mechanisms of acute vascular rejection and cell-mediated rejection are poorly understood. METHODS: Using a heterotopic rat-to-mouse cardiac transplantation model, we demonstrate that IL-12p40 attenuates both allogeneic and xenogeneic acute vascular rejection pathology by suppressing B-cell activation and anti-rat isotype switching. To study the mechanism of xenogeneic cell-mediated rejection, we use B-cell deficient mice that only develop cell-mediated rejection pathology. To elucidate the role of IL-12 in cell-mediated rejection, we generated B cell/ IL-12p40 double knockout mice. RESULTS: We demonstrate that xenogeneic cell-mediated rejection is mediated by CD4+ T cells, and is accompanied by elevated FasL and granzyme mRNA expression. Strikingly, by generating B cell/IL-12p40 double knockout mice, we demonstrate that xenogeneic cell-mediated rejection is IL-12p40 dependent. In contrast, we demonstrate that allogeneic cellular rejection is IL-12p40 independent. CONCLUSIONS: We conclude that IL-12 plays a dual role in xenotransplantation by driving xenogeneic CD4+ T cell responses but suppressing both allogeneic and xenogeneic B cell responses. Therefore, the mechanism of allogeneic and xenogeneic transplantation rejection is differentially regulated by IL-12.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Graft Rejection/immunology , Interleukin-12/immunology , Protein Subunits/immunology , Animals , Antibodies, Heterophile/biosynthesis , Base Sequence , DNA, Complementary/genetics , Graft Rejection/pathology , Graft Rejection/prevention & control , Heart Transplantation/immunology , Heart Transplantation/pathology , Humans , Immune Tolerance , Interleukin-12/deficiency , Interleukin-12/genetics , Interleukin-12 Subunit p40 , Lymphocyte Activation , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Protein Subunits/deficiency , Protein Subunits/genetics , Rats , Rats, Inbred Lew , Transplantation, Heterologous , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL