Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Cell Death Differ ; 28(5): 1705-1719, 2021 05.
Article in English | MEDLINE | ID: mdl-33288903

ABSTRACT

Foxo1 transcription factor is an evolutionarily conserved regulator of cell metabolism, oxidative stress, inflammation, and apoptosis. Activation of Hedgehog/Gli signaling is known to regulate cell growth, differentiation, and immune function. However, the molecular mechanisms by which interactive cell signaling networks restrain oxidative stress response and necroptosis are still poorly understood. Here, we report that myeloid-specific Foxo1 knockout (Foxo1M-KO) mice were resistant to oxidative stress-induced hepatocellular damage with reduced macrophage/neutrophil infiltration, and proinflammatory mediators in liver ischemia/reperfusion injury (IRI). Foxo1M-KO enhanced ß-catenin-mediated Gli1/Snail activity, and reduced receptor-interacting protein kinase 3 (RIPK3) and NIMA-related kinase 7 (NEK7)/NLRP3 expression in IR-stressed livers. Disruption of Gli1 in Foxo1M-KO livers deteriorated liver function, diminished Snail, and augmented RIPK3 and NEK7/NLRP3. Mechanistically, macrophage Foxo1 and ß-catenin colocalized in the nucleus, whereby the Foxo1 competed with T-cell factor (TCF) for interaction with ß-catenin under inflammatory conditions. Disruption of the Foxo1-ß-catenin axis by Foxo1 deletion enhanced ß-catenin/TCF binding, activated Gli1/Snail signaling, leading to inhibited RIPK3 and NEK7/NLRP3. Furthermore, macrophage Gli1 or Snail knockout activated RIPK3 and increased hepatocyte necroptosis, while macrophage RIPK3 ablation diminished NEK7/NLRP3-driven inflammatory response. Our findings underscore a novel molecular mechanism of the myeloid Foxo1-ß-catenin axis in regulating Hedgehog/Gli1 function that is key in oxidative stress-induced liver inflammation and necroptosis.


Subject(s)
Forkhead Box Protein O1/metabolism , Hedgehog Proteins/metabolism , Inflammasomes/metabolism , beta Catenin/metabolism , Animals , Humans , Mice , Oxidative Stress
2.
Liver Transpl ; 26(1): 113-126, 2020 01.
Article in English | MEDLINE | ID: mdl-31642174

ABSTRACT

Tissue inhibitor of metalloproteinase (TIMP) 3 is a naturally occurring inhibitor of a broad range of proteases, with key roles in extracellular matrix turnover and in the pathogenesis of various diseases. In this study, we investigated the response of mice lacking TIMP3 (TIMP3-/-) to hepatic ischemia/reperfusion injury (IRI). We report here that TIMP3-/- mice showed an enhanced inflammatory response, exacerbated organ damage, and further impaired liver function after IRI when compared with their wild-type littermates. Loss of TIMP3 led to the cleavage and shedding of E-cadherin during hepatic IRI; the full-length 120-kDa E-cadherin and the ratio of 38-kDa C-terminal fragment/120-kDa E-cadherin were decreased and increased, respectively, in TIMP3-/- livers after IRI. Moreover, GI254023X, a potent inhibitor of a disintegrin and metalloprotease (ADAM) 10, was capable of partially rescuing the expression of E-cadherin in the TIMP3-null hepatocytes. The proteolysis of E-cadherin in the TIMP3-/- livers was also linked to the loss of ß-catenin from the hepatocyte membranes and to an increased susceptibility to apoptosis after liver IRI. In a similar fashion, depression of the E-cadherin/ß-catenin complex mediated by TIMP3 deletion and knockdown of ß-catenin by small interfering RNA were both capable of inducing caspase activation in isolated hepatocytes subjected to H2 O2 oxidative stress. Hence, these results support a protective role for TIMP3 expression in sheltering the hepatocyte E-cadherin/ß-catenin complex from proteolytic processing and inhibiting apoptosis after hepatic IRI.


Subject(s)
Liver Transplantation , Reperfusion Injury , Animals , Cadherins , Hepatocytes , Ischemia , Liver , Metalloproteases , Mice , Tissue Inhibitor of Metalloproteinase-3/genetics , beta Catenin
3.
Cell Mol Immunol ; 17(12): 1245-1256, 2020 12.
Article in English | MEDLINE | ID: mdl-31673056

ABSTRACT

Notch signaling plays important roles in the regulation of immune cell functioning during the inflammatory response. Activation of the innate immune signaling receptor NLRP3 promotes inflammation in injured tissue. However, it remains unknown whether Jagged1 (JAG1)-mediated myeloid Notch1 signaling regulates NLRP3 function in acute liver injury. Here, we report that myeloid Notch1 signaling regulates the NLRP3-driven inflammatory response in ischemia/reperfusion (IR)-induced liver injury. In a mouse model of liver IR injury, Notch1-proficient (Notch1FL/FL) mice receiving recombinant JAG1 showed a reduction in IR-induced liver injury and increased Notch intracellular domain (NICD) and heat shock transcription factor 1 (HSF1) expression, whereas myeloid-specific Notch1 knockout (Notch1M-KO) aggravated hepatocellular damage even with concomitant JAG1 treatment. Compared to JAG1-treated Notch1FL/FL controls, Notch1M-KO mice showed diminished HSF1 and Snail activity but augmented NLRP3/caspase-1 activity in ischemic liver. The disruption of HSF1 reduced Snail activation and enhanced NLRP3 activation, while the adoptive transfer of HSF1-expressing macrophages to Notch1M-KO mice augmented Snail activation and mitigated IR-triggered liver inflammation. Moreover, the knockdown of Snail in JAG1-treated Notch1FL/FL livers worsened hepatocellular functioning, reduced TRX1 expression and increased TXNIP/NLRP3 expression. Ablation of myeloid Notch1 or Snail increased ASK1 activation and hepatocellular apoptosis, whereas the activation of Snail increased TRX1 expression and reduced TXNIP, NLRP3/caspase-1, and ROS production. Our findings demonstrated that JAG1-mediated myeloid Notch1 signaling promotes HSF1 and Snail activation, which in turn inhibits NLRP3 function and hepatocellular apoptosis leading to the alleviation of IR-induced liver injury. Hence, the Notch1/HSF1/Snail signaling axis represents a novel regulator of and a potential therapeutic target for liver inflammatory injury.


Subject(s)
Heat Shock Transcription Factors/metabolism , Inflammasomes/metabolism , Inflammation/pathology , Jagged-1 Protein/metabolism , Liver/injuries , Myeloid Cells/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Receptor, Notch1/metabolism , Animals , Apoptosis , Carrier Proteins/metabolism , Immunity , Liver/metabolism , Liver/pathology , Macrophages/metabolism , Macrophages/pathology , Mice, Knockout , Models, Biological , Necrosis , Neutrophils/metabolism , Neutrophils/pathology , Reperfusion Injury/pathology , Signal Transduction , Snail Family Transcription Factors/metabolism , Thioredoxins/metabolism
4.
Liver Transpl ; 25(2): 288-301, 2019 02.
Article in English | MEDLINE | ID: mdl-30358115

ABSTRACT

The purpose of this study was to assess the significance of tenascin-C (Tnc) expression in steatotic liver ischemia/reperfusion injury (IRI). The critical shortage in donor organs has led to the use of steatotic livers in transplantation regardless of their elevated susceptibility to hepatic IRI. Tnc is an endogenous danger signal extracellular matrix molecule involved in various aspects of immunity and tissue injury. In the current study, mice were fed with a steatosis-inducing diet and developed approximately 50% hepatic steatosis, predominantly macrovesicular, before being subjected to hepatic IRI. We report here that lipid accumulation in hepatocytes inflated the production of Tnc in steatotic livers and in isolated hepatic stellate cells. Moreover, we show that the inability of Tnc-/- deficient steatotic mice to express Tnc significantly protected these mice from liver IRI. Compared with fatty controls, Tnc-/- steatotic mice showed significantly reduced serum transaminase levels and enhanced liver histological preservation at both 6 and 24 hours after hepatic IRI. The lack of Tnc expression resulted in impaired lymphocyte antigen 6 complex, locus (Ly6G) neutrophil and macrophage antigen-1 (Mac-1) leukocyte recruitment as well as in decreased expression of proinflammatory mediators (interleukin 1ß, tumor necrosis factor α, and chemokine [C-X-C motif] ligand 2) after liver reperfusion. Myeloperoxidase (MPO) is the most abundant cytotoxic enzyme secreted by neutrophils and a key mediator of neutrophil-induced oxidative tissue injuries. Using an in vitro model of steatosis, we also show that Tnc markedly potentiated the effect of steatotic hepatocytes on neutrophil-derived MPO activity. In conclusion, our data support the view that inhibition of Tnc is a promising therapeutic approach to lessen inflammation in steatotic livers and to maximize their successful use in organ transplantation.


Subject(s)
Allografts/pathology , Liver Transplantation/adverse effects , Liver/pathology , Non-alcoholic Fatty Liver Disease/pathology , Reperfusion Injury/pathology , Tenascin/metabolism , Allografts/cytology , Allografts/metabolism , Animals , Disease Models, Animal , Donor Selection/standards , Extracellular Matrix/pathology , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Inflammation Mediators/metabolism , Lipid Metabolism , Liver/cytology , Liver/metabolism , Liver Function Tests , Liver Transplantation/standards , Male , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/etiology , Reperfusion Injury/etiology , Tenascin/genetics
5.
Am J Pathol ; 188(8): 1820-1832, 2018 08.
Article in English | MEDLINE | ID: mdl-29870740

ABSTRACT

Matrix metalloproteinase-9 (MMP-9) is abundantly expressed by infiltrating leukocytes and contributes to the pathogenesis of hepatic ischemia and reperfusion injury (IRI). On the other hand, its physiological inhibitor, the tissue inhibitor of metalloproteinases-1 (TIMP-1), is available in insufficient levels to hamper MMP-9 activity during hepatic IRI. In this study, we generated recombinant adeno-associated virus type 8 vectors (rAAV8) encoding mouse TIMP-1 driven by a liver-specific thyroxine-binding globulin promoter as a strategy to increase the levels of TIMP-1 during liver IRI. Biodistribution analysis confirmed selective overexpression of TIMP-1 in livers of rAAV8-TIMP-1 vector treated C57BL/6 mice. rAAV8-TIMP-1-treated mice showed reduced MMP-9 activity, diminished leukocyte trafficking and activation, lowered transaminase levels, and improved histology after liver IRI. Moreover, the rAAV8-TIMP-1 vector therapy enhanced significantly the 7-day survival rate of TIMP-1-/- mice subjected to hepatic IRI. Neutrophils are the first cells recruited to inflamed tissues and, once activated, they release nuclear DNA-forming web-like structures, known as neutrophil extracellular traps. It was found that TIMP-1 has the ability to reduce formation of neutrophil extracellular traps and, consequently, limit the impact of neutrophil extracellular trap-mediated cytotoxicity in hepatic IRI. This is the first report demonstrating that TIMP-1 overexpression is hepatoprotective in ischemia and reperfusion injury. Hence, TIMP-1 may represent a promising molecule for drug development to treat liver IRI.


Subject(s)
Dependovirus/genetics , Extracellular Traps , Liver Diseases/prevention & control , Reperfusion Injury/prevention & control , Tissue Inhibitor of Metalloproteinase-1/genetics , Animals , Cells, Cultured , Leukocytes/metabolism , Leukocytes/pathology , Liver Diseases/genetics , Liver Diseases/pathology , Mice , Mice, Inbred C57BL , Neutrophils/metabolism , Neutrophils/pathology , Reperfusion Injury/genetics , Reperfusion Injury/pathology
6.
PLoS One ; 10(9): e0137642, 2015.
Article in English | MEDLINE | ID: mdl-26355684

ABSTRACT

Hepatic ischemia and reperfusion injury (IRI) is an inflammatory condition and a significant cause of morbidity and mortality after surgery. Matrix metalloproteinases (MMPs) have been widely implicated in the pathogenesis of inflammatory diseases. Among the different MMPs, gelatinases (MMP-2 and MMP-9) are within the most prominent MMPs detected during liver IRI. While the role of MMP-9 in liver damage has been fairly documented, direct evidence of the role for MMP-2 activity in hepatic IRI remains to be established. Due to the lack of suitable inhibitors to target individual MMPs in vivo, gene manipulation is as an essential tool to assess MMP direct contribution to liver injury. Hence, we used MMP-2-/- deficient mice and MMP-2+/+ wild-type littermates to examine the function of MMP-2 activity in hepatic IRI. MMP-2 expression was detected along the sinusoids of wild-type livers before and after surgery and in a small population of leukocytes post-IRI. Compared to MMP-2+/+ mice, MMP-2 null (MMP-2-/-) mice showed exacerbated liver damage at 6, 24, and 48 hours post-reperfusion, which was fatal in some cases. MMP-2 deficiency resulted in upregulation of MMP-9 activity, spontaneous leukocyte infiltration in naïve livers, and amplified MMP-9-dependent transmigration of leukocytes in vitro and after hepatic IRI. Moreover, complete loss of MMP-2 activity impaired the degradation of poly (ADP-ribose) polymerase (PARP-1) in extensively damaged livers post-reperfusion. However, the administration of a PARP-1 inhibitor to MMP-2 null mice restored liver preservation to almost comparable levels of MMP-2+/+ mice post-IRI. Deficient PARP-1 degradation in MMP-2-null sinusoidal endothelial cells correlated with their increased cytotoxicity, evaluated by the measurement of LDH efflux in the medium. In conclusion, our results show for the first time that MMP-2 gene deletion exacerbates liver IRI. Moreover, they offer new insights into the MMP-2 modulation of inflammatory responses, which could be relevant for the design of new pharmacological MMP-targeted agents to treat hepatic IRI.


Subject(s)
Gene Deletion , Liver/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Cell Movement/genetics , Cell Movement/immunology , Cytokines/metabolism , Disease Models, Animal , Enzyme Activation , Gene Expression , Inflammation Mediators/metabolism , Leukocyte Count , Leukocytes/immunology , Leukocytes/metabolism , Leukocytes/pathology , Liver/blood supply , Liver/pathology , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase Inhibitors/pharmacology , Mice , Mice, Knockout , Poly (ADP-Ribose) Polymerase-1 , Proteolysis , Reperfusion Injury/mortality , Reperfusion Injury/pathology
7.
Matrix Biol ; 44-46: 147-56, 2015.
Article in English | MEDLINE | ID: mdl-25599939

ABSTRACT

The liver is a large highly vascularized organ with a central function in metabolic homeostasis, detoxification, and immunity. Due to its roles, the liver is frequently exposed to various insults which can cause cell death and hepatic dysfunction. Alternatively, the liver has a remarkable ability to self-repair and regenerate after injury. Liver injury and regeneration have both been linked to complex extracellular matrix (ECM) related pathways. While normal degradation of ECM components is an important feature of tissue repair and remodeling, irregular ECM turnover contributes to a variety of liver diseases. Matrix metalloproteinases (MMPs) are the main enzymes implicated in ECM degradation. MMPs not only remodel the ECM, but also regulate immune responses. In this review, we highlight some of the MMP-attributed roles in acute and chronic liver injury and emphasize the need for further experimentation to better understand their functions during hepatic physiological conditions and disease progression.


Subject(s)
Liver Diseases/pathology , Liver/enzymology , Matrix Metalloproteinases/metabolism , Extracellular Matrix/metabolism , Humans , Liver/pathology , Liver Diseases/enzymology , Signal Transduction , Tissue Inhibitor of Metalloproteinases/metabolism , Wound Healing
8.
PLoS One ; 9(5): e96913, 2014.
Article in English | MEDLINE | ID: mdl-24819536

ABSTRACT

Cyclooxygenase-2 (COX-2) is a mediator of hepatic ischemia and reperfusion injury (IRI). While both global COX-2 deletion and pharmacologic COX-2 inhibition ameliorate liver IRI, the clinical use of COX-2 inhibitors has been linked to increased risks of heart attack and stroke. Therefore, a better understanding of the role of COX-2 in different cell types may lead to improved therapeutic strategies for hepatic IRI. Macrophages of myeloid origin are currently considered to be important sources of the COX-2 in damaged livers. Here, we used a Cox-2flox conditional knockout mouse (COX-2-M/-M) to examine the function of COX-2 expression in myeloid cells during liver IRI. COX-2-M/-M mice and their WT control littermates were subjected to partial liver ischemia followed by reperfusion. COX-2-M/-M macrophages did not express COX-2 upon lipopolysaccharide stimulation and COX-2-M/-M livers showed reduced levels of COX-2 protein post-IRI. Nevertheless, selective deletion of myeloid cell-derived COX-2 failed to ameliorate liver IRI; serum transaminases and histology were comparable in both COX-2-M/-M and WT mice. COX-2-M/-M livers, like WT livers, developed extensive necrosis, vascular congestion, leukocyte infiltration and matrix metalloproteinase-9 (MMP-9) expression post-reperfusion. In addition, myeloid COX-2 deletion led to a transient increase in IL-6 levels after hepatic reperfusion, when compared to controls. Administration of celecoxib, a selective COX-2 inhibitor, resulted in significantly improved liver function and histology in both COX-2-M/-M and WT mice post-reperfusion, providing evidence that COX-2-mediated liver IRI is caused by COX-2 derived from a source(s) other than myeloid cells. In conclusion, these results support the view that myeloid COX-2, including myeloid-macrophage COX-2, is not responsible for the hepatic IRI phenotype.


Subject(s)
Cyclooxygenase 2/metabolism , Liver Diseases/enzymology , Myeloid Cells/enzymology , Reperfusion Injury/enzymology , Animals , Cyclooxygenase 2/genetics , Macrophages/metabolism , Mice
9.
J Hepatol ; 60(5): 1032-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24412604

ABSTRACT

BACKGROUND & AIMS: Organ shortage has led to the use of steatotic livers in transplantation, despite their elevated susceptibility to ischemia/reperfusion injury (IRI). Matrix metalloproteinase-9 (MMP-9), an inducible gelatinase, is emerging as a central mediator of leukocyte traffic into inflamed tissues. However, its role in steatotic hepatic IRI has yet to be demonstrated. METHODS: We examined the function of MMP-9 in mice fed with a high-fat diet (HFD), which developed approximately 50% hepatic steatosis, predominantly macrovesicular, prior to partial hepatic IRI. RESULTS: The inability of MMP-9(-/-) deficient steatotic mice to express MMP-9 significantly protected these mice from liver IRI. Compared to fatty controls, MMP-9(-/-) steatotic livers showed significantly reduced leukocyte infiltration, proinflammatory cytokine expression, and liver necrosis. Loss of MMP-9 activity preserved platelet endothelial cell adhesion molecule-1 (PECAM-1) expression, a modulator of vascular integrity at the endothelial cell-cell junctions in steatotic livers after IRI. Using in vitro approaches, we show that targeted inhibition of MMP-9 sheltered the extracellular portion of PECAM-1 from proteolytic processing, and disrupted leukocyte migration across this junctional molecule. Moreover, the evaluation of distinct parameters of regeneration, proliferating cell nuclear antigen (PCNA) and histone H3 phosphorylation (pH3), provided evidence that hepatocyte progression into S phase and mitosis was notably enhanced in MMP-9(-/-) steatotic livers after IRI. CONCLUSIONS: MMP-9 activity disrupts vascular integrity at least partially through a PECAM-1 dependent mechanism and interferes with regeneration of steatotic livers after IRI. Our novel findings establish MMP-9 as an important mediator of steatotic liver IRI.


Subject(s)
Liver Regeneration/physiology , Matrix Metalloproteinase 9/deficiency , Non-alcoholic Fatty Liver Disease/physiopathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Reperfusion Injury/physiopathology , Animals , Diet, High-Fat/adverse effects , Endothelium, Vascular/metabolism , Leukocytes/pathology , Leukocytes/physiology , Liver/pathology , Liver/physiopathology , Liver Regeneration/genetics , Male , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/pathology , Reperfusion Injury/pathology
10.
Hepatology ; 56(3): 1074-85, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22407827

ABSTRACT

UNLABELLED: Hepatic ischemia and reperfusion injury (IRI) remains an important challenge in clinical orthotopic liver transplantation (OLT). Tissue inhibitor of metalloproteinase-1 (TIMP-1) is the major endogenous regulator of matrix metalloproteinase-9 (MMP-9). In this study we investigated the functional significance of TIMP-1 expression in a well-established mouse model of partial liver IRI. Compared to wildtype mice, TIMP-1(-/-) mice showed further impaired liver function and histological preservation after IRI. Notably, TIMP-1 deficiency led to lethal liver IRI, as over 60% of the TIMP-1(-/-) mice died postreperfusion, whereas all TIMP-1(+/+) mice recovered and survived surgery. Lack of TIMP-1 expression was accompanied by markedly high levels of MMP-9 activity, which facilitates leukocyte transmigration across vascular barriers in hepatic IRI. Indeed, TIMP-1(-/-) livers were characterized by massive leukocyte infiltration and by up-regulation of proinflammatory mediators, including tumor necrosis factor alpha, interferon-gamma, and inducible nitric oxide synthase post-IRI. The inability of TIMP-1(-/-) mice to express TIMP-1 increased the levels of active caspase-3 and depressed the expression of Bcl-2 and the phosphorylation of Akt, emphasizing an important role for TIMP-1 expression on hepatocyte survival. Using independent parameters of regeneration, 5-bromodeoxyuridine incorporation, proliferating cell nuclear antigen expression, and histone H3 phosphorylation, we provide evidence that hepatocyte progression into S phase and mitosis was impaired in TIMP-1-deficient livers after IRI. Inhibition of the cell cycle progression by TIMP-1 deficiency was linked to depressed levels of cyclins-D1 and -E and to a disrupted c-Met signaling pathway, as evidenced by reduced phosphorylated c-Met expression and elevated c-Met ectodomain shedding postliver IRI. CONCLUSION: These results support a critical protective function for TIMP-1 expression on promoting survival and proliferation of liver cells and on regulating leukocyte recruitment and activation in liver IRI.


Subject(s)
Ischemia/etiology , Liver/blood supply , Reperfusion Injury/etiology , Tissue Inhibitor of Metalloproteinase-1/deficiency , Animals , Ischemia/mortality , Mice , Mice, Inbred C57BL , Reperfusion Injury/mortality
11.
J Am Coll Surg ; 214(4): 505-15; discussion 515-6, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22321520

ABSTRACT

BACKGROUND: Ischemia-reperfusion injury (IRI) can occur during hepatic surgery and transplantation. IRI causes hepatic mitochondrial and microcirculatory impairment, resulting in acute liver dysfunction and failure. We proposed a novel strategy of regulated hepatic reperfusion (RHR) to reverse the cellular metabolic deficit that incurred during organ ischemia by using a substrate-enriched, oxygen-saturated, and leukocyte-depleted perfusate delivered under regulated reperfusion pressure, temperature, and pH. We investigate the use of RHR in mitigating IRI after a prolonged period of warm ischemia. METHODS: Using a 2-hour liver warm ischemia swine model, 2 methods of liver reperfusion were compared. The control group (n = 6) received conventional reperfusion with unmodified portal venous blood under unregulated reperfusion pressure, temperature, and pH. The experimental group (n = 6) received RHR. We analyzed the effects of RHR on post-reperfusion hemodynamic changes, liver function, and 7-day animal survival. RESULTS: RHR resulted in 100% survival compared with 50% in the control group (p = 0.05). Post-reperfusion syndrome was not observed in the RHR group, but it occurred in 83% of the control group. RHR resulted in a lesser degree of change from baseline serum alanine aminotransferase levels, aspartate aminotransferase, and lactate dehydrogenase after reperfusion compared with the control group. Histopathologic evaluation showed minimal ischemic changes in the RHR group, whereas a considerable degree of coagulative hepatocellular necrosis was observed in the control group. CONCLUSIONS: Regulated hepatic reperfusion mitigates IRI, facilitates liver function recovery, and improves survival after a prolonged period of hepatic warm ischemia. This novel strategy has potential applicability to clinical hepatic surgery and liver transplantation when marginal grafts are used.


Subject(s)
Liver/surgery , Reperfusion Injury/prevention & control , Reperfusion/methods , Warm Ischemia , Animals , Disease Models, Animal , Kaplan-Meier Estimate , Liver/pathology , Liver/physiology , Liver Function Tests , Liver Transplantation , Pilot Projects , Reperfusion/adverse effects , Resuscitation , Survival Rate , Swine , Time Factors , Warm Ischemia/mortality
12.
Hepatology ; 54(6): 2125-36, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21898491

ABSTRACT

Hepatic ischemia/reperfusion (IRI) injury remains a major challenge in clinical orthotopic liver transplantation (OLT). Tenascin-C (Tnc) is an extracellular matrix protein (ECM) involved in various aspects of immunity and tissue injury. Using a Tnc-deficient mouse model, we present data that suggest an active role for Tnc in liver IRI. We show that Tnc-deficient mice have a reduction in liver damage and a significant improvement in liver regeneration after IRI. The inability of Tnc(-/-) mice to express Tnc significantly reduced the levels of active caspase-3/transferase-mediated dUTP nick end-labeling (TUNEL) apoptotic markers and enhanced the expression of the proliferation cell nuclear antigen (PCNA) after liver IRI. The lack of Tnc expression resulted in impaired leukocyte recruitment and decreased expressions of interleukin (IL)-1ß, IL-6, and CXCL2 after liver reperfusion. Tnc-deficient livers were characterized by altered expression patterns of vascular adhesion molecules, such as vascular cell adhesion molecule-1 and platelet endothelial cell adhesion molecule-1 post-IRI. Moreover, matrix metalloproteinase-9 (MMP-9) synthesis, which facilitates leukocyte transmigration across vascular barriers in liver IRI, was markedly down-regulated in the absence of Tnc. We also show that Tnc is capable of inducing MMP-9 expression in isolated neutrophils through Toll-like receptor 4. Therefore, our data suggest that Tnc is a relevant mediator of the pathogenic events underlying liver IRI. The data also support the view that studies aimed at further understanding how newly synthesized ECM molecules, such as Tnc, participate in inflammatory responses are needed to improve therapeutic approaches in liver IRI.


Subject(s)
Liver Regeneration/physiology , Reperfusion Injury/immunology , Tenascin/deficiency , Animals , Caspase 3/metabolism , In Situ Nick-End Labeling , Intercellular Adhesion Molecule-1/biosynthesis , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , Liver/pathology , Liver/physiology , Matrix Metalloproteinase 9/biosynthesis , Mice , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Tenascin/immunology , Toll-Like Receptor 4/physiology , Vascular Cell Adhesion Molecule-1/biosynthesis
13.
Curr Opin Organ Transplant ; 16(1): 34-40, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21150609

ABSTRACT

PURPOSE OF REVIEW: Hepatic ischemia reperfusion injury (IRI) linked to leukocyte recruitment and subsequent release of cytokines and free radicals remains a significant complication in organ transplantation. The aim of this review is to bring attention to advances made in our understanding of the mechanisms of leukocyte recruitment to sites of inflammatory stimulation in liver IRI. RECENT FINDINGS: Leukocyte transmigration across endothelial and extracellular matrix barriers is dependent on adhesive events, as well as on focal matrix degradation mechanisms. Whereas adhesion molecules are critical for the successful promotion of leukocyte transmigration by providing leukocyte attachment to the vascular endothelium, matrix metalloproteinases (MMPs) are important for facilitating leukocyte movement across vascular barriers. Among different MMPs, MMP-9, an inducible gelatinase expressed by leukocytes during hepatic IRI, is emerging as an important mediator of leukocyte traffic to inflamed liver. SUMMARY: It is generally accepted that the understanding of the molecular mechanisms involved in leukocyte recruitment will lead to the development of novel targeted therapeutic approaches for hepatic IRI and liver transplantation. Here, we review mechanisms of leukocyte traffic in liver IRI and the role of some of the proteins that are thought to be important for this process.


Subject(s)
Endothelium, Vascular/pathology , Extracellular Matrix Proteins/metabolism , Leukocytes/pathology , Liver/blood supply , Reperfusion Injury/pathology , Endothelium, Vascular/metabolism , Humans , Leukocytes/metabolism , Liver/metabolism , Liver/pathology , Matrix Metalloproteinase 9/metabolism , Reperfusion Injury/metabolism
14.
Am J Pathol ; 174(6): 2265-77, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19443702

ABSTRACT

Matrix metalloproteinase 9 (MMP-9) is a critical mediator of leukocyte migration in hepatic ischemia/reperfusion (I/R) injury. To test the relevance of inducible nitric oxide synthase (iNOS) expression on the regulation of MMP-9 activity in liver I/R injury, our experiments included both iNOS-deficient mice and mice treated with ONO-1714, a specific iNOS inhibitor. The inability of iNOS-deficient mice to generate iNOS-derived nitric oxide (NO) profoundly inhibited MMP-9 activity and depressed leukocyte migration in livers after I/R injury. While macrophages expressed both iNOS and MMP-9 in damaged wild-type livers, neutrophils expressed MMP-9 and were virtually negative for iNOS; however, exposure of isolated murine neutrophils and macrophages to exogenous NO increased MMP-9 activity in both cell types, suggesting that NO may activate MMP-9 in leukocytes by either autocrine or paracrine mechanisms. Furthermore, macrophage NO production through the induction of iNOS was capable of promoting neutrophil transmigration across fibronectin in a MMP-9-dependent manner. iNOS expression in liver I/R injury was also linked to liver apoptosis, which was reduced in the absence of MMP-9. These results suggest that MMP-9 activity induced by iNOS-derived NO may also lead to detachment of hepatocytes from the extracellular matrix and cell death, in addition to regulating leukocyte migration across extracellular matrix barriers. These data provide evidence for a novel mechanism by which MMP-9 can mediate iNOS-induced liver I/R injury.


Subject(s)
Chemotaxis, Leukocyte/physiology , Liver/immunology , Matrix Metalloproteinase 9/metabolism , Nitric Oxide Synthase Type II/metabolism , Reperfusion Injury/immunology , Animals , Apoptosis/physiology , Blotting, Western , Immunohistochemistry , In Situ Nick-End Labeling , Liver/injuries , Liver/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Nitric Oxide/immunology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/deficiency , Reperfusion Injury/metabolism
15.
J Immunol ; 180(3): 1843-53, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18209082

ABSTRACT

Cyclooxygenase-2 (COX-2) is a prostanoid-synthesizing enzyme that is critically implicated in a variety of pathophysiological processes. Using a COX-2-deficient mouse model, we present data that suggest that COX-2 has an active role in liver ischemia/reperfusion (I/R) injury. We demonstrate that COX-2-deficient mice had a significant reduction in liver damage after I/R insult. The inability of COX-2(-/-) to elaborate COX-2 products favored a Th2-type response in these mice. COX-2(-/-) livers after I/R injury showed significantly decreased levels of IL-2, as well as IL-12, a cytokine known to have a central role in Th1 effector cell differentiation. Moreover, such livers expressed enhanced levels of the anti-inflammatory cytokine IL-10, shifting the balance in favor of a Th2 response in COX-2-deficient mice. The lack of COX-2 expression resulted in decreased levels of CXCL2, a neutrophil-activating chemokine, reduced infiltration of MMP-9-positive neutrophils, and impaired late macrophage activation in livers after I/R injury. Additionally, Bcl-2 and Bcl-x(L) were normally expressed in COX-2(-/-) livers after injury, whereas respective wild-type controls were almost depleted of these two inhibitors of cell death. In contrast, caspase-3 activation and TUNEL-positive cells were depressed in COX-2(-/-) livers. Therefore, our data support the concept that COX-2 is involved in the pathogenic events occurring in liver I/R injury. The data also suggest that potential valuable therapeutic approaches in liver I/R injury may result from further studies aimed at identifying specific COX-2-derived prostanoid pathways.


Subject(s)
Cyclooxygenase 2/deficiency , Liver/blood supply , Liver/immunology , Neutrophil Infiltration , Reperfusion Injury/immunology , Th2 Cells/immunology , Animals , Celecoxib , Chemokine CXCL2/metabolism , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/genetics , Cyclooxygenase 2 Inhibitors/administration & dosage , Cytokines/genetics , Cytokines/metabolism , Down-Regulation , Liver/enzymology , Macrophage Activation , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors , Mice , Mice, Knockout , Neutrophil Infiltration/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyrazoles/administration & dosage , Reperfusion Injury/enzymology , Reperfusion Injury/prevention & control , Sulfonamides/administration & dosage , Th1 Cells/immunology , bcl-X Protein/metabolism
16.
Hepatology ; 47(1): 186-98, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17880014

ABSTRACT

UNLABELLED: Leukocyte transmigration across endothelial and extracellular matrix protein barriers is dependent on adhesion and focal matrix degradation events. In the present study we investigated the role of metalloproteinase-9 (MMP-9/gelatinase B) in liver ischemia/reperfusion (I/R) injury using MMP-9-deficient (MMP-9(-/-)) animals and mice treated with a specific anti-MMP-9 neutralizing antibody or with a broad gelatinase inhibitor for both MMP-9 and metalloproteinase-2 (MMP-2/gelatinase A). Compared to wild-type mice, MMP-9(-/-) mice and mice treated with an anti-MMP-9 antibody showed significantly reduced liver damage. In contrast, mice treated with a broad gelatinase inhibitor showed rather inferior protection against I/R injury and were characterized by persistent ongoing liver inflammation, suggesting that MMP-2 and MMP-9 may have distinct roles in this type of injury. MMP-9 was mostly detected in Ly-6G and macrophage antigen-1 leukocytes adherent to the vessel walls and infiltrating the damaged livers of wild-type mice after liver I/R injury. Leukocyte traffic and cytokine expression were markedly impaired in livers of MMP-9(-/-) animals and in livers of mice treated with anti-MMP-9 antibody after I/R injury; however, initiation of the endothelial adhesion cascades was similar in both MMP-9(-/-) and control livers. We also showed that MMP-9-specific inhibition disrupted neutrophil migration across fibronectin in transwell filters and depressed myeloperoxidase (MPO) activation in vitro. CONCLUSION: These results support critical functions for MMP-9 in leukocyte recruitment and activation leading to liver damage. Moreover, they provide the rationale for identifying inhibitors to specifically target MMP-9 in vivo as a potential therapeutic approach in liver I/R injury.


Subject(s)
Liver Diseases/enzymology , Matrix Metalloproteinase 9/deficiency , Matrix Metalloproteinase Inhibitors , Reperfusion Injury/enzymology , Animals , Antibodies/therapeutic use , Cell Movement/physiology , Enzyme Inhibitors/therapeutic use , Fibronectins/physiology , Intercellular Adhesion Molecule-1/metabolism , Leukocytes/physiology , Liver Diseases/immunology , Liver Diseases/pathology , Liver Diseases/prevention & control , Male , Matrix Metalloproteinase 9/immunology , Mice , Mice, Knockout , Neutrophils/physiology , Peroxidase/metabolism , Reperfusion Injury/immunology , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Vascular Cell Adhesion Molecule-1/metabolism , Warm Ischemia/adverse effects
17.
Am J Pathol ; 170(2): 567-77, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17255325

ABSTRACT

Ischemia/reperfusion injury is a major cause of the highly dysfunctional rate observed in marginal steatotic orthotopic liver transplantation. In this study, we document that the interactions between fibronectin, a key extracellular matrix protein, and its integrin receptor alpha4beta1, expressed on leukocytes, specifically up-regulated the expression and activation of metalloproteinase-9 (MMP-9, gelatinase B) in a well-established steatotic rat liver model of ex vivo ice-cold ischemia followed by isotransplantation. The presence of the active form of MMP-9 was accompanied by massive intragraft leukocyte infiltration, high levels of proinflammatory cytokines, such as interleukin-1beta and tumor necrosis factor-alpha, and impaired liver function. Interestingly, MMP-9 activity in steatotic liver grafts was, to a certain extent, independent of the expression of its natural inhibitor, the tissue inhibitor of metalloproteinases-1. Moreover, the blockade of fibronectin-alpha4beta1-integrin interactions inhibited the expression/activation of MMP-9 in steatotic orthotopic liver transplantations without significantly affecting the expression of metalloproteinase-2 (MMP-2, gelatinase A). Finally, we identified T lymphocytes and monocytes/macrophages as major sources of MMP-9 in steatotic liver grafts. Hence, these findings reveal a novel aspect of the function of fibronectin-alpha4beta1 integrin interactions that holds significance for the successful use of marginal steatotic livers in transplantation.


Subject(s)
Fatty Liver/enzymology , Gene Expression Regulation, Enzymologic , Integrin alpha4beta1/metabolism , Matrix Metalloproteinase 9/biosynthesis , Reperfusion Injury/enzymology , Up-Regulation , Animals , Cold Ischemia , Fatty Liver/pathology , Interleukin-1beta/biosynthesis , Liver/enzymology , Liver/pathology , Liver Transplantation , Macrophages/metabolism , Macrophages/pathology , Male , Matrix Metalloproteinase 2/biosynthesis , Rats , Rats, Sprague-Dawley , Rats, Zucker , Reperfusion Injury/pathology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Transplantation, Isogeneic , Tumor Necrosis Factor-alpha/biosynthesis
18.
Am J Pathol ; 162(4): 1229-39, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12651615

ABSTRACT

We tested a hypothesis that interactions between fibronectin (FN), the major extracellular matrix component, and its integrin alpha 4 beta 1 receptor is important in the development of ischemia/reperfusion injury of steatotic liver transplants. We examined the effect of connecting segment-1 (CS1) peptide-facilitated blockade of FN-alpha 4 beta 1 interaction in a well-established steatotic rat liver model of ex vivo cold ischemia followed by iso-transplantation. In this model, CS1 peptides were administered through the portal vein of steatotic Zucker rat livers before and after cold ischemic storage. Lean Zucker recipients of fatty liver transplants received an additional 3-day course of CS1 peptides after transplant. CS1 peptide therapy significantly inhibited the recruitment of T lymphocytes, neutrophil activation/infiltration, and repressed the expression of proinflammatory tumor necrosis factor-alpha and interferon-gamma. Moreover, it resulted in selective inhibition of inducible nitric oxide synthase expression, peroxynitrite formation, and hepatic necrosis. Importantly, CS1 peptide therapy improved function/histological preservation of steatotic liver grafts, and extended their 14-day survival in lean recipients from 40% in untreated to 100% in CS1-treated OLTs. Thus, CS1 peptide-mediated blockade of FN-alpha 4 beta 1 interaction protects against severe ischemia/reperfusion injury experienced otherwise by steatotic OLTs. These novel findings document the potential of targeting FN-alpha 4 beta 1 in vivo interaction to increase the transplant donor pool through modulation of marginal steatotic livers.


Subject(s)
Integrin alpha4beta1/antagonists & inhibitors , Liver Circulation/physiology , Liver Transplantation/pathology , Obesity/genetics , Peptide Fragments/pharmacology , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Animals , Immunohistochemistry , Integrin alpha4beta1/chemistry , Male , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Obesity/pathology , Peroxidase/metabolism , Rats , Rats, Sprague-Dawley , Rats, Zucker , Reverse Transcriptase Polymerase Chain Reaction
19.
J Am Soc Nephrol ; 14(3): 745-54, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12595512

ABSTRACT

Heme oxygenase-1 (HO-1) overexpression using gene transfer protects rat livers against ischemia/reperfusion (I/R) injury. This study evaluates the effects of Ad-HO-1 gene transfer in a rat renal isograft model. Donor LEW kidneys were perfused with Ad-HO-1, Ad-beta-gal, or PBS, stored at 4 degrees C for 24 h, and transplanted orthotopically into LEW recipients, followed by contralateral native nephrectomy. Serum creatinine, urine protein/creatinine ratios, severity of histologic changes, HO-1 mRNA/protein expression, and HO enzymatic activity were analyzed. Ad-HO-1 gene transfer conferred a survival advantage when compared with PBS- and Ad-beta-gal-treated controls, with median survival of 100, 7, and 7 d, respectively (P < 0.01). Serum creatinine levels were elevated at day 7 in all groups (range, 2.2 to 5.8 mg/dl) but recovered to 1.0 mg/dl by day 14 (P < 0.01) in Ad-HO-1 group, which was sustained thereafter. Urine protein/creatinine ratio at day 7 was elevated in both PBS and Ad-beta-gal, as compared with the Ad-HO-1 group (12.0 and 9.8 versus 5.0; P < 0.005); histologically, ATN and glomerulosclerosis was more severe in Ad-beta-gal group at all time points. Reverse transcriptase-PCR-based HO-1 gene expression was significantly increased before reperfusion (P < 0.001) and remained increased in the Ad-HO-1-treated group for 3 d after transplantation. Concomitantly, HO enzymatic activity was increased at transplantation and at 3 d posttransplant in the Ad-HO-1 group, compared with Ad-beta-gal controls (P < 0.05); tubular HO-1 expression was discernible early posttransplant in the Ad-HO-1 group alone. These findings are consistent with protective effects of HO-1 overexpression using a gene transfer approach against severe renal I/R injury, with reduced mortality and attenuation of tissue injury.


Subject(s)
Graft Survival/physiology , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Kidney Transplantation , Reperfusion Injury/metabolism , Reperfusion Injury/therapy , Adenoviridae/genetics , Animals , Cold Temperature , Gene Expression Regulation, Enzymologic , Gene Transfer Techniques , Heme Oxygenase-1 , Immunohistochemistry , Kidney/enzymology , Kidney/pathology , Kidney Tubular Necrosis, Acute/metabolism , Kidney Tubular Necrosis, Acute/pathology , Kidney Tubular Necrosis, Acute/therapy , Male , Rats , Rats, Inbred Lew , Reperfusion Injury/pathology , Reverse Transcriptase Polymerase Chain Reaction , Severity of Illness Index
20.
Am J Transplant ; 2(9): 843-9, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12392290

ABSTRACT

Ischemia and reperfusion (IR) injury remains a significant problem in clinical liver transplantation. We investigated the effects of lymphocyte depletion with FTY720 in models of warm hepatic IR. Using 60-min partial warm hepatic IR, three groups of rats were studied: Sham--laparotomy alone; Control--water p.o. x 3 d before ischemia; Treatment--FTY720 p.o. x 3 d before ischemia. Animals were sacrificed for analysis at 6 h and 24 h post reperfusion. The effect of FTY720 pretreatment on survival was also studied using 150 min total hepatic IR with portojugular shunt. FTY720 treatment significantly reduced serum glutamic pyruvic transaminase and peripheral blood lymphocytes compared to controls at 6h and 24h (p < 0.0005). Histological grade was significantly improved in treated livers vs. controls (p < 0.05). CD3 immunocytochemical analysis revealed a significant reduction in T-cell infiltration in FTY720-treated livers (p < 0.0002). No difference in tissue myeloperoxidase levels was observed. Seven-day survival was significantly improved in treated rats vs. controls following total hepatic ischemia (p < 0.05). In conclusion, FTY720 ameliorates the biochemical and histological manifestations of hepatic IR by preventing T-lymphocyte infiltration and prolongs survival following a more severe ischemic insult. Myeloperoxidase data suggest this mechanism is independent of neutrophil activation. These results indicate that T lymphocytes are pivotal mediators in hepatic IR and may have important implications in liver transplantation.


Subject(s)
Immunosuppressive Agents/pharmacology , Liver/drug effects , Propylene Glycols/pharmacology , Reperfusion Injury/prevention & control , T-Lymphocytes/drug effects , Animals , Cell Movement/drug effects , Cell Movement/immunology , Fingolimod Hydrochloride , Immunohistochemistry , Liver/immunology , Male , Rats , Rats, Inbred Lew , Reperfusion Injury/immunology , Sphingosine/analogs & derivatives , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...