151
|
Hui W, Jinxiang Z, Heshui W, Zhuoya L, Qichang Z. Bone marrow and non-bone marrow TLR4 regulates hepatic ischemia/reperfusion injury. Biochem Biophys Res Commun 2009; 389:328-32. [PMID: 19723506 DOI: 10.1016/j.bbrc.2009.08.149] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Accepted: 08/26/2009] [Indexed: 01/04/2023]
Abstract
Hepatic ischemia-reperfusion injury (IRI) is a highly coordinated process often observed during liver transplantation, liver surgery, and hemorrhagic shock. Signaling through toll-like receptor 4 (TLR4), which is widely expressed on all kinds of liver cells, appears critical in the pathogenesis of IRI. Although the role of TLR4 expressed on non-parenchymal cells (NPCs) of the liver, including Kupffer cells and neutrophils, in IRI has been widely studied, TLR4 signaling on liver sinusoidal endothelial cells (LSECs) or hepatocytes in the process of IRI, and their coordination with bone marrow derived TLR4 in the late reperfusion stage, is largely unknown. We produced TLR4 chimeric mice that received hepatic IRI, and examined the degree of liver injury and the underlying mechanisms of injury. Results indicated that mutation of TLR4 on bone-marrow or non-bone marrow derived cells reduced hepatic IRI in the late reperfusion stage via cytokine release and neutrophil infiltration, while non-bone marrow derived TLR4 regulated the expression of ICAM-1 on hepatocytes and LSECs, exacerbating their injury. In conclusion, both TLR4 on bone marrow derived and non-bone marrow derived cells were necessary in the process of hepatic IRI.
Collapse
Affiliation(s)
- Wang Hui
- Department of Medical Genetics, Tongji Medical College Affiliated to Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | | | | | | | | |
Collapse
|
152
|
Uchida Y, Freitas MCS, Zhao D, Busuttil RW, Kupiec-Weglinski JW. The inhibition of neutrophil elastase ameliorates mouse liver damage due to ischemia and reperfusion. Liver Transpl 2009; 15:939-47. [PMID: 19642132 PMCID: PMC2752954 DOI: 10.1002/lt.21770] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neutrophils are considered crucial effector cells in the pathophysiology of organ ischemia/reperfusion injury (IRI). Although neutrophil elastase (NE) accounts for a substantial portion of the neutrophil activity, the function of NE in liver IRI remains unclear. This study focuses on the role of NE in the mechanism of liver IRI. Partial warm ischemia was produced in the left and middle hepatic lobes of C57BL/6 mice for 90 minutes, and this was followed by 6 to 24 hours of reperfusion. Mice were treated with neutrophil elastase inhibitor (NEI; 2 mg/kg per os) at 60 minutes prior to the ischemia insult. NEI treatment significantly reduced serum alanine aminotransferase levels in comparison with controls. Histological examination of liver sections revealed that unlike in controls, NEI treatment ameliorated hepatocellular damage and decreased local neutrophil infiltration, as assessed by myeloperoxidase assay, naphthol AS-D chloroacetate esterase stains, and immunohistochemistry (anti-Ly-6G). The expression of pro-inflammatory cytokines (tumor necrosis factor alpha and interleukin 6) and chemokines [chemokine (C-X-C motif) ligand 1 (CXCL-1), CXCL-2, and CXCL-10] was significantly reduced in the NEI treatment group, along with diminished apoptosis, according to terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling staining and caspase-3 activity. In addition, toll-like receptor 4 (TLR4) expression was diminished in NEI-pretreated livers, and this implies a putative role of NE in the TLR4 signal transduction pathway. Thus, targeting NE represents a useful approach for preventing liver IRI and hence expanding the organ donor pool and improving the overall success of liver transplantation. Liver Transpl 15:939-947, 2009. (c) 2009 AASLD.
Collapse
Affiliation(s)
- Yoichiro Uchida
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | | | | | | | | |
Collapse
|
153
|
Shaw PJ, Ganey PE, Roth RA. Trovafloxacin enhances the inflammatory response to a Gram-negative or a Gram-positive bacterial stimulus, resulting in neutrophil-dependent liver injury in mice. J Pharmacol Exp Ther 2009; 330:72-8. [PMID: 19351866 PMCID: PMC2700160 DOI: 10.1124/jpet.109.151068] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2009] [Accepted: 04/06/2009] [Indexed: 01/22/2023] Open
Abstract
Trovafloxacin (TVX), a fluoroquinolone antibiotic, has been strongly linked with several cases of idiosyncratic hepatotoxicity in humans. Previous studies showed that a modest inflammatory stress induced by a Gram-negative bacterial stimulus [i.e., lipopolysaccharide (LPS)] rendered nontoxic doses of TVX hepatotoxic in mice. This study compared the interaction of TVX with Gram-negative and Gram-positive stimuli. Mice were given TVX 3 h before LPS (Gram-negative stimulus) or a peptidoglycan-lipoteichoic acid (PGN-LTA) mixture isolated from Staphylococcus aureus (Gram-positive stimulus). Administration of TVX, LPS, or PGN-LTA alone was nonhepatotoxic. However, TVX administration before PGN-LTA or LPS resulted in significant liver injury that occurred with similar time courses. TVX/PGN-LTA-induced hepatocellular necrosis was primarily localized to centrilobular regions, whereas that caused by TVX/LPS was predominantly midzonal. Administration of either LPS or PGN-LTA alone led to increased plasma concentrations of several cytokines and chemokines at a time near the onset of liver injury. TVX administration before LPS enhanced the concentrations of all of these cytokines, whereas TVX treatment before PGN-LTA increased all of the cytokines except tumor necrosis factor (TNF)-alpha and interferon-gamma. Liver injury was reduced in TVX/LPS- and TVX/PGN-LTA-treated mice given an antibody to CD18 and also in mice deficient in neutrophil [polymorphonuclear neutrophil (PMN)] elastase. Hepatic PMN accumulation and TNF-alpha production after TVX/PGN-LTA-, but not after TVX/LPS-coexposure, was CD18-dependent. In summary, TVX significantly enhanced the murine inflammatory response to either a Gram-negative or a Gram-positive stimulus and caused hepatotoxicity that developed similarly and was dependent on PMN activation in mice but that differed in lesion location and cytokine profile.
Collapse
Affiliation(s)
- Patrick J Shaw
- Department of Pharmacology and Toxicology, National Food Safety and Toxicology Center, Michigan State University, East Lansing, MI 48824, USA
| | | | | |
Collapse
|
154
|
Hamada T, Duarte S, Tsuchihashi S, Busuttil RW, Coito AJ. Inducible nitric oxide synthase deficiency impairs matrix metalloproteinase-9 activity and disrupts leukocyte migration in hepatic ischemia/reperfusion injury. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 174:2265-77. [PMID: 19443702 DOI: 10.2353/ajpath.2009.080872] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
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.
Collapse
Affiliation(s)
- Takashi Hamada
- The Dumont-UCLA Transplant Center, 77-120 CHS, Box: 957054, Los Angeles, CA 90095-7054, USA
| | | | | | | | | |
Collapse
|
155
|
Song H, Bergstrasser C, Rafat N, Höger S, Schmidt M, Endres N, Goebeler M, Hillebrands JL, Brigelius-Flohé R, Banning A, Beck G, Loesel R, Yard BA. The carbon monoxide releasing molecule (CORM-3) inhibits expression of vascular cell adhesion molecule-1 and E-selectin independently of haem oxygenase-1 expression. Br J Pharmacol 2009; 157:769-80. [PMID: 19422386 DOI: 10.1111/j.1476-5381.2009.00215.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND AND PURPOSE Although carbon monoxide (CO) can modulate inflammatory processes, the influence of CO on adhesion molecules is less clear. This might be due to the limited amount of CO generated by haem degradation. We therefore tested the ability of a CO releasing molecule (CORM-3), used in supra-physiological concentrations, to modulate the expression of vascular cell adhesion molecule (VCAM)-1 and E-selectin on endothelial cells and the mechanism(s) involved. EXPERIMENTAL APPROACH Human umbilical vein endothelial cells (HUVECs) were stimulated with tumour necrosis factor (TNF)-alpha in the presence or absence of CORM-3. The influence of CORM-3 on VCAM-1 and E-selectin expression and the nuclear factor (NF)-kappaB pathway was assessed by flow cytometry, Western blotting and electrophoretic mobility shift assay. KEY RESULTS CORM-3 inhibited the expression of VCAM-1 and E-selectin on TNF-alpha-stimulated HUVEC. VCAM-1 expression was also inhibited when CORM-3 was added 24 h after TNF-alpha stimulation or when TNF-alpha was removed. This was paralleled by deactivation of NF-kappaB and a reduction in VCAM-1 mRNA. Although TNF-alpha removal was more effective in this regard, VCAM-1 protein was down-regulated more rapidly when CORM-3 was added. CORM-3 induced haem oxygenase-1 (HO-1) in a dose- and time-dependent manner, mediated by the transcription factor, Nrf2. CORM-3 was still able to down-regulate VCAM-1 expression in HUVEC transfected with siRNA for HO-1 or Nrf2. CONCLUSIONS AND IMPLICATIONS Down-regulation of VCAM and E-selectin expression induced by CORM-3 was independent of HO-1 up-regulation and was predominantly due to inhibition of sustained NF-kappaB activation.
Collapse
Affiliation(s)
- H Song
- Vth Medical Department, University Hospital Mannheim, Mannheim, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
156
|
Affiliation(s)
- Srinivasan Dasarathy
- Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA.
| |
Collapse
|
157
|
Kuriyama N, Isaji S, Hamada T, Kishiwada M, Ohsawa I, Usui M, Sakurai H, Tabata M, Suzuki K, Uemoto S. Activated protein C prevents hepatic ischaemia-reperfusion injury in rats. Liver Int 2009; 29:299-307. [PMID: 18507760 DOI: 10.1111/j.1478-3231.2008.01796.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Hepatic ischaemia-reperfusion injury (IRI) is a serious complication of liver surgery, especially extended hepatectomy and liver transplantation. Activated protein C (APC), a potent anticoagulant serine protease, has been shown to have cell-protective properties by virtue of its anti-inflammatory and anti-apoptotic activities. METHODS The present study was designed to examine the cytoprotective effects of APC in a 60-min warm-IRI rat model. RESULTS Following a single intravenous injection of APC before reperfusion, APC exerted cytoprotective effects 4 h after reperfusion, as evidenced by: (i) decreased levels of transaminase and improved histological findings of IRI, (ii) reduced infiltration and activation of neutrophils, macrophages and T cells, (iii) reduced expression of tumour necrosis factor-alpha, (iv) reduced expression of P-selectin and intracellular adhesion molecule-1, (v) inhibited coagulation and attenuated sinusoidal endothelial cell injury, (vi) improved hepatic microcirculation and (vii) decreased transferase-mediated dUTP nick end-labelling-positive cells. These effects of APC were observed 4 h but not 24 h after reperfusion. However, multiple injections of APC after reperfusion significantly decreased the levels of transaminase and the activity of myeloperoxidase, and improved histological findings of IRI 24 h after reperfusion. CONCLUSION These results suggest that APC is a promising therapeutic option for hepatic warm-IRI; however, multiple injections of APC are necessary to maintain its cell-protective action over the long term.
Collapse
Affiliation(s)
- Naohisa Kuriyama
- Department of Hepatobiliary Pancreatic Surgery, Mie University Graduate School of Medicine, Mie, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
158
|
Antoniades CG, Berry PA, Wendon JA, Vergani D. The importance of immune dysfunction in determining outcome in acute liver failure. J Hepatol 2008; 49:845-61. [PMID: 18801592 DOI: 10.1016/j.jhep.2008.08.009] [Citation(s) in RCA: 266] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Acute liver failure (ALF) shares striking similarities with septic shock with regard to the features of systemic inflammation, progression to multiple organ dysfunction and functional immunoparesis. While the existence of opposing systemic pro- and anti-inflammatory profiles resulting in organ failure and immune dysfunction are well recognised in septic shock, characterization of these processes in ALF has only recently been described. This review explores the evolution of the systemic inflammation in acute liver failure, its relation to disease progression, exacerbation of liver injury and development of innate immune dysfunction and extra-hepatic organ failure as sequelae. Defects in innate immunity are described in hepatic and extra-hepatic compartments. Clinical studies measuring levels of pro- and anti-inflammatory cytokines and expression of the antigen presentation molecule HLA-DR on monocytes, in combination with ex-vivo experiments, demonstrate that the persistence of a compensatory anti-inflammatory response syndrome, leading to functional monocyte deactivation, is a central event in the evolution of systemic immune dysfunction. Accurate immune profiling in ALF may permit the development of immunomodulatory strategies in order to improve outcome in this condition.
Collapse
|
159
|
Everson CA, Thalacker CD, Hogg N. Phagocyte migration and cellular stress induced in liver, lung, and intestine during sleep loss and sleep recovery. Am J Physiol Regul Integr Comp Physiol 2008; 295:R2067-74. [PMID: 18945949 DOI: 10.1152/ajpregu.90623.2008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Sleep is understood to possess recuperative properties and, conversely, sleep loss is associated with disease and shortened life span. Despite these critical attributes, the mechanisms and functions by which sleep and sleep loss impact health still are speculative. One of the most consistent, if largely overlooked, signs of sleep loss in both humans and laboratory rats is a progressive increase in circulating phagocytic cells, mainly neutrophils. The destination, if any, of the increased circulating populations has been unknown and, therefore, its medical significance has been uncertain. The purpose of the present experiment was to determine the content and location of neutrophils in liver and lung tissue of sleep-deprived rats. These are two principal sites affected by neutrophil migration during systemic inflammatory illness. The content of neutrophils in the intestine also was determined. Sleep deprivation in rats was produced for 5 and 10 days by the Bergmann-Rechtschaffen disk method, which has been validated for its high selectivity under freely moving conditions and which was tolerated and accompanied by a deep negative energy balance. Comparison groups included basal conditions and 48 h of sleep recovery after 10 days of sleep loss. Myeloperoxidase (MPO), an enzyme constituent of neutrophils, was extracted from liver, lung, and intestinal tissues, and its activity was determined by spectrophotometry. Leukocytes were located in vasculature and interstitial spaces in the liver and the lung by immunohistochemistry. Heme oxygenase-1, also known as heat shock protein-32 and a marker of cellular stress, and corticosterone also were measured. The results indicate neutrophil migration into extravascular liver and lung tissue concurrent with cell stress and consistent with tissue injury or infection induced by sleep loss. Plasma corticosterone was unchanged. Recovery sleep was marked by increased lung heme oxygenase-1, increased intestinal MPO activity, and abnormally low corticosterone, suggesting ongoing reactive processes as a result of prior sleep deprivation.
Collapse
Affiliation(s)
- Carol A Everson
- Department of Neurology, The Medical College of Wisconsin, Neurology Research 151, VAMC, 5000 West National Ave., Milwaukee, WI 53295, USA.
| | | | | |
Collapse
|
160
|
Ramaiah SK, Jaeschke H. Hepatic Neutrophil Infiltration in the Pathogenesis of Alcohol-Induced Liver Injury. Toxicol Mech Methods 2008; 17:431-40. [DOI: 10.1080/00952990701407702] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
161
|
Pevni D, Frolkis I, Schwartz D, Schwartz I, Chernichovski T, Kramer A, Ben-Gal Y, Uretzky G, Shapira I, Weinbroum A. New evidence for the role of TNF-alpha in liver ischaemic/reperfusion injury. Eur J Clin Invest 2008; 38:649-55. [PMID: 18837741 DOI: 10.1111/j.1365-2362.2008.01996.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Tumour necrosis factor-alpha (TNF-alpha) plays a key role in causing ischaemia/reperfusion (I/R) injury. I/R also causes activation of xanthine oxidase and dehydrogenase (XDH + XO) system that, via generated free radicals, causes organ damage. We investigated the effect of ischaemia, reperfusion and non-ischaemic prolonged perfusion (NIP) on TNF-alpha and XDH + XO production in an isolated perfused rat liver model. MATERIALS AND METHODS Rat livers underwent 150 min NIP (control group) or two hours of ischaemia followed by reperfusion (I/R group). TNF-alpha (TNF-alpha mRNA and protein level), XDH + XO production and bile secretion were determined in tissue and effluent at baseline, at 120 min of ischaemia, after 30 min of reperfusion (I/R group) and after 120 and 150 min of prolonged perfusion (control). RESULTS Unexpectedly, neither ischaemia nor reperfusion had any effect on TNF-alpha production. TNF-alpha in effluent was 11 +/- 4.8 pg mL(-1) at baseline, 7 +/- 3.2 pg mL(-1) at the end of ischaemia, and 13 +/- 5.3 pg mL(-1) after 30 min of reperfusion. NIP, however, caused a significant increase of TNF-alpha synthesis and release. TNF-alpha effluent level after 120 and 150 min of perfusion was 392 +/- 78.7 pg mL(-1) and 408 +/- 64.3 pg mL(-1), respectively. TNF-alpha mRNA in tissue was also significantly elevated compared to baseline levels (1.31 +/- 0.2 P < 0.001 and 1.38 P < 0.002, respectively). Decrease of liver function (expressed by bile secretion) during I/R and NIP was accompanied by significant XDH + XO elevation. CONCLUSION This is the first evidence that NIP, and not I/R, is the decisive trigger for TNF-alpha production. This study leads to a better understanding of pathogenesis of liver I/R and perfusion damage.
Collapse
Affiliation(s)
- D Pevni
- Department of Cardiac Surgery, Tel-Aviv Sourasky Medical Centre, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | | | | | | | | | | | | | | | | |
Collapse
|
162
|
Pro-inflammatory response and adverse drug reactions: Mechanisms of action of ximelagatran on chemokine and cytokine activation in a monocyte in vitro model. Toxicol In Vitro 2008; 22:1588-94. [DOI: 10.1016/j.tiv.2008.06.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2007] [Revised: 05/15/2008] [Accepted: 06/24/2008] [Indexed: 02/08/2023]
|
163
|
Banerjee A, Lee JH, Ramaiah SK. Interaction of osteopontin with neutrophil alpha(4)beta(1) and alpha(9)beta(1) integrins in a rodent model of alcoholic liver disease. Toxicol Appl Pharmacol 2008; 233:238-46. [PMID: 18778724 DOI: 10.1016/j.taap.2008.08.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2008] [Revised: 07/24/2008] [Accepted: 08/05/2008] [Indexed: 12/17/2022]
Abstract
Previous studies from our laboratory have reported that osteopontin (OPN) mediated higher hepatic neutrophil infiltration makes female rats more susceptible to alcoholic steatohepatitis (ASH) than their male counterparts. The objective of the current work was to investigate the patho-mechanism by which OPN attracts the hepatic neutrophils in ASH. We hypothesized that OPN-mediated hepatic neutrophil infiltration is a result of signaling by N-terminal integrin binding motif (SLAYGLR) of OPN through its receptor alpha(9)beta(1) (VLA9) and alpha(4)beta(1) (VLA4) integrins on neutrophils. Compared to the males, females in the ASH group exhibited higher expression of alpha(4)beta(1) and alpha(9)beta(1) protein and mRNA and a significant decrease in the expression of these integrins was observed in rats treated with neutralizing OPN antibody. Immunoprecipitation experiments suggested the binding of OPN to alpha(4)beta(1) and alpha(9)beta(1) integrins. OPN-mediated neutrophil infiltration was also confirmed using Boyden chamber assays, and antibodies directed against alpha(4) and beta(1) integrins was found to significantly inhibit neutrophilic migration in vitro. In conclusion, these data suggest that SLAYGLR-mediated alpha(4)beta(1) and alpha(9)beta(1) integrin signaling may be responsible for higher hepatic neutrophil infiltration and higher liver injury in the rat ASH model.
Collapse
Affiliation(s)
- Atrayee Banerjee
- Department of Toxicology & Pathobiology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX 77843-4467, USA
| | | | | |
Collapse
|
164
|
LEUKOTRIENE B4/LEUKOTRIENE B4 RECEPTOR PATHWAY IS INVOLVED IN HEPATIC MICROCIRCULATORY DYSFUNCTION ELICITED BY ENDOTOXIN. Shock 2008; 30:87-91. [DOI: 10.1097/shk.0b013e31815d06a1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
165
|
Xu L, Fang H, Frucht DM. Anthrax lethal toxin increases superoxide production in murine neutrophils via differential effects on MAPK signaling pathways. THE JOURNAL OF IMMUNOLOGY 2008; 180:4139-47. [PMID: 18322225 DOI: 10.4049/jimmunol.180.6.4139] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The combination of lethal factor and its receptor-binding partner, protective Ag, is termed lethal toxin (LT) and has critical pathogenic activity during infection with Bacillus anthracis. We herein report that anthrax LT binds and enters murine neutrophils, leading to the cleavage of mitogen-activated protein kinase kinase/MEK/MAPKK 1-4 and 6, but not mitogen-activated protein kinase kinase 5 and 7. Anthrax LT treatment of neutrophils disrupts signaling to downstream MAPK targets in response to TLR stimulation. Following anthrax LT treatment, ERK family and p38 phosphorylation are nearly completely blocked, but signaling to JNK family members persists in vitro and ex vivo. In contrast to previous reports involving human neutrophils, anthrax LT treatment of murine neutrophils increases their production of superoxide in response to PMA or TLR stimulation in vitro or ex vivo. Although this enhanced superoxide production correlates with effects due to the LT-induced blockade of ERK signaling, it requires JNK signaling that remains largely intact despite the activity of anthrax LT. These findings reveal a previously unrecognized mechanism through which anthrax LT supports a critical proinflammatory response of murine neutrophils.
Collapse
Affiliation(s)
- Lixin Xu
- Division of Monoclonal Antibodies, Office of Biotechnology Products, Office of Pharmaceutical Science, Center for Drug Evaluation and Research, US Food and Drug Administration, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
166
|
Abstract
Portal hypertension and its complications account for the majority of morbidity and mortality that occurs in patients with cirrhosis. In addition to portal hypertension, a number of other vascular syndromes are also of great importance, especially the ischemia-reperfusion (IR) injury. With the identification of major vascular defects that could account for many of the clinical sequelae of these syndromes, the liver vasculature field has now integrated very closely with the broader vascular biology discipline. In that spirit, the Henry and Lillian Stratton Basic Research Single Topic Conference was held on the topic of Vascular Biology and Pathobiology of the Liver. The course took place approximately 10 years after the first American Association for the Study of Liver Disease (AASLD)-sponsored conference on this topic that occurred in Reston, Virginia. The conference initiated with an introduction to basic vascular cell signaling and then explored vascular biology specifically as it relates to liver cells. Subsequently, specific disease syndromes were discussed in more detail including portal hypertension and IR injury. Finally, clinical and translational sessions focused on emerging therapies and technologies to treat vascular diseases of the liver.
Collapse
Affiliation(s)
- Yasuko Iwakiri
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Matthew Grisham
- Department of Molecular & Cellular Physiology, LSU Health Sciences Center, Shreveport, Louisiana 71130, USA
| | - Vijay Shah
- GI Research Unit and Fitterman Center for Digestive Disease, Mayo Clinic, Rochester, Minnesota 55905, USA
| |
Collapse
|
167
|
Cervantes-Sandoval I, Serrano-Luna JDJ, García-Latorre E, Tsutsumi V, Shibayama M. Characterization of brain inflammation during primary amoebic meningoencephalitis. Parasitol Int 2008; 57:307-13. [PMID: 18374627 DOI: 10.1016/j.parint.2008.01.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2007] [Revised: 01/21/2008] [Accepted: 01/26/2008] [Indexed: 01/21/2023]
Abstract
Naegleria fowleri is a free-living amoeba and the etiologic agent of primary amoebic meningoencephalitis (PAM). Trophozoites reach the brain by penetrating the olfactory epithelium, and invasion of the olfactory bulbs results in an intense inflammatory reaction. The contribution of the inflammatory response to brain damage in experimental PAM has not been delineated. Using both optical and electron microscopy, we analyzed the morphologic changes in the brain parenchyma due to inflammation during experimental PAM. Several N. fowleri trophozoites were observed in the olfactory bulbs 72 h post-inoculation, and the number of amoebae increased rapidly over the next 24 h. Eosinophils and neutrophils surrounding the amoebae were then noted at later times during infection. Electron microscopic examination of the increased numbers of neutrophils and the interactions with trophozoites indicated an active attempt to eliminate the amoebae. The extent of inflammation increased over time, with a predominant neutrophil response indicating important signs of damage and necrosis of the parenchyma. These data suggest a probable role of inflammation in tissue damage. To test the former hypothesis, we used CD38-/- knockout mice with deficiencies in chemotaxis to compare the rate of mortality with the parental strain, C57BL/6J. The results showed that inflammation and mortality were delayed in the knockout mice. Based on these results, we suggest that the host inflammatory response and polymorphonuclear cell lysis contribute to a great extent to the central nervous system tissue damage.
Collapse
Affiliation(s)
- Isaac Cervantes-Sandoval
- Department of Experimental Pathology, Center for Research and Advanced Studies, National Polytechnic Institute, Mexico City, Mexico
| | | | | | | | | |
Collapse
|
168
|
Abstract
Liver damage leads to an inflammatory response and to the activation and proliferation of mesenchymal cell populations within the liver which remodel the extracellular matrix as part of an orchestrated wound-healing response. Chronic damage results in a progressive accumulation of scarring proteins (fibrosis) that, with increasing severity, alters tissue structure and function, leading to cirrhosis and liver failure. Efforts to modulate the fibrogenesis process have focused on understanding the biology of the heterogeneous liver fibroblast populations. The fibroblasts are derived from sources within and out with the liver. Fibroblasts expressing alpha-smooth muscle actin (myofibroblasts) may be derived from the transdifferentiation of quiescent hepatic stellate cells. Other fibroblasts emerge from the portal tracts within the liver. At least a proportion of these cells in diseased liver originate from the bone marrow. In addition, fibrogenic fibroblasts may also be generated through liver epithelial (hepatocyte and biliary epithelial cell)-mesenchymal transition. Whatever their origin, it is clear that fibrogenic fibroblast activity is sensitive to (and may be active in) the cytokine and chemokine profiles of liver-resident leucocytes such as macrophages. They may also be a component driving the regeneration of tissue. Understanding the complex intercellular interactions regulating liver fibrogenesis is of increasing importance in view of predicted increases in chronic liver disease and the current paucity of effective therapies.
Collapse
|
169
|
Chen LC, Laskin JD, Gordon MK, Laskin DL. Regulation of TREM expression in hepatic macrophages and endothelial cells during acute endotoxemia. Exp Mol Pathol 2008; 84:145-55. [PMID: 18222421 PMCID: PMC2752215 DOI: 10.1016/j.yexmp.2007.11.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2007] [Accepted: 11/15/2007] [Indexed: 01/13/2023]
Abstract
Triggering receptor expressed on myeloid cells (TREM) regulates inflammatory responses to lipopolysaccharide (LPS). In these studies, we analyzed the expression of TREM in hepatic macrophages and endothelial cells which play a central role in LPS clearance. LPS administration to C3H/HeOuJ mice resulted in a rapid induction of TREM-1 and TREM-3, but a decrease in TREM-2 in liver macrophages and endothelial cells. The observation that TREM family members are detectable in endothelial cells is novel and demonstrates that their expression is not limited to myeloid cells. LPS-induced alterations in TREM expression were not evident in cells from C3H/HeJ TLR-4 mutant mice, indicating that the response is dependent on TLR-4. IL-1beta and TNFalpha upregulated TREM-1 and TREM-3 expression and suppressed TREM-2 expression in macrophages and endothelial cells. This activity involved PI3-kinase and p38 MAP kinase signaling. Interestingly, no significant differences were noted in TREM expression between wild-type and TNFR1-/- mice treated with LPS. Treatment of macrophages and endothelial cells with LPS upregulated expression of nitric oxide synthase-2 (NOS-2). This was blocked by TREM-1 Fc/fusion protein, indicating that TREM-1 mediates LPS-induced NOS-2 expression. These results suggest that TREM proteins are important in the inflammatory response of hepatic macrophages and endothelial cells to acute endotoxemia.
Collapse
Affiliation(s)
- Li C Chen
- Rutgers University, Piscataway, New Jersey 08854, USA
| | | | | | | |
Collapse
|
170
|
Hamada T, Tsuchihashi S, Avanesyan A, Duarte S, Moore C, Busuttil RW, Coito AJ. Cyclooxygenase-2 deficiency enhances Th2 immune responses and impairs neutrophil recruitment in hepatic ischemia/reperfusion injury. THE JOURNAL OF IMMUNOLOGY 2008; 180:1843-53. [PMID: 18209082 DOI: 10.4049/jimmunol.180.3.1843] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
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.
Collapse
Affiliation(s)
- Takashi Hamada
- The Dumont-University of California Los Angeles Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | |
Collapse
|
171
|
Swee M, Wilson CL, Wang Y, McGuire JK, Parks WC. Matrix metalloproteinase-7 (matrilysin) controls neutrophil egress by generating chemokine gradients. J Leukoc Biol 2008; 83:1404-12. [PMID: 18334539 DOI: 10.1189/jlb.0108016] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Matrilysin [matrix metalloproteinase 7 (MMP7)] is induced by mucosal injury of many tissues. To assess function of this proteinase, we subjected wild-type and Mmp7(-/-) mice to acute colon injury. When matrilysin expression was increasing, 73% of wild-type mice died, whereas only 32% of Mmp7(-/-) mice succumbed. Although re-epithelialization was delayed in Mmp7(-/-) mice, overall injury did not differ markedly between genotypes. We hypothesized that differences in acute inflammation caused increased mortality in wild-type mice. Indeed, whereas overall neutrophil influx into tissue was similar in wild-type and Mmp7(-/-) mice, their location and extent of migration differed between genotypes. Neutrophils were dispersed throughout the mucosa and within the lumen of wild-type mice, but these leukocytes were largely confined to the submucosa in Mmp7(-/-) mice. The levels of neutrophil chemokines, keratinocyte-derived chemokine and MIP-2, increased in the colon tissue of both genotypes, but these factors were detected only in lumenal lavages of wild-type mice. Our findings indicate that matrilysin mediates beneficial and deleterious effects in response to injury. On one hand, it promotes re-epithelialization, but it also controls the transepithelial influx of neutrophils, which if excessive, can lead to tissue damage.
Collapse
Affiliation(s)
- Mei Swee
- Center for Lung Biology and Institute for Stem Cell and Regenerative Medicine and Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | | | | | | | | |
Collapse
|
172
|
Kim SY, Weinstein DA, Starost MF, Mansfield BC, Chou JY. Necrotic foci, elevated chemokines and infiltrating neutrophils in the liver of glycogen storage disease type Ia. J Hepatol 2008; 48:479-85. [PMID: 18191274 PMCID: PMC2744599 DOI: 10.1016/j.jhep.2007.11.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2007] [Revised: 10/24/2007] [Accepted: 11/03/2007] [Indexed: 12/25/2022]
Abstract
BACKGROUND/AIMS Glycogen storage disease type Ia (GSD-Ia) patients manifest the long-term complication of hepatocellular adenoma (HCA) of unknown etiology. We showed previously that GSD-Ia mice exhibit neutrophilia and elevated serum cytokine levels. This study was conducted to evaluate whether human GSD-Ia patients exhibit analogous increases and whether in GSD-Ia mice a correlation exists between immune abnormalities and, biochemical and histological alterations in the liver. METHODS Differential leukocyte counts and cytokine levels were investigated in GSD-Ia patients. Hepatic chemokine production, neutrophil infiltration, and histological abnormalities were investigated in GSD-Ia mice. RESULTS We show that GSD-Ia patients exhibit increased peripheral neutrophil counts and serum interleukin-8 (IL-8). Compared to normal subjects, HCA-bearing GSD-Ia patients have a 2.8-fold higher serum IL-8 concentration, while GSD-Ia patients without HCA have a 1.4-fold higher concentration. Hepatic injury in GSD-Ia mice is evidenced by necrotic foci, markedly elevated infiltrating neutrophils, and increased hepatic production of chemokines. CONCLUSIONS Peripheral neutrophilia and elevated serum chemokines are characteristic of GSD-Ia with HCA-bearing GSD-Ia patients having the highest serum IL-8. In GSD-Ia mice these elevations correlate with elevated hepatic chemokine levels, neutrophil infiltration, and necrosis. Taken together, peripheral neutrophilia and increased serum chemokines may indicate hepatic injuries in GSD-Ia.
Collapse
Affiliation(s)
- So Youn Kim
- Section on Cellular Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - David A. Weinstein
- Glycogen Storage Disease Program, Division of Pediatric Endocrinology, University of Florida College of Medicine, Gainesville, FL 32610
| | - Matthew F. Starost
- Division of Veterinary Resources, National Institutes of Health, Bethesda, MD 20892
| | - Brian C. Mansfield
- Section on Cellular Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - Janice Y. Chou
- Section on Cellular Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
173
|
Analysis of chemokines and reactive oxygen species formation by rat and human neutrophils induced by microcystin-LA, -YR and -LR. Toxicon 2008; 51:1274-80. [PMID: 18405933 DOI: 10.1016/j.toxicon.2008.02.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2007] [Revised: 02/15/2008] [Accepted: 02/20/2008] [Indexed: 11/21/2022]
Abstract
Microcystins (MC), a family of heptapeptide toxins produced by some genera of Cyanobacteria, have potent hepatotoxicity and tumor-promoting activity. Leukocyte infiltration in the liver was observed in MC-induced acute intoxication. Although the mechanisms of hepatotoxicity are still unclear, neutrophil infiltration in the liver may play an important role in triggering toxic injury and tumor development. The present study reports the effects of MC-LA, MC-YR and MC-LR (1 and 1000 nM) on human and rat neutrophils functions in vitro. Cell viability, DNA fragmentation, mitochondrial membrane depolarization and intracellular reactive oxygen species (ROS) levels were measured by flow cytometry. Extracellular ROS content was measured by lucigenin-amplified chemiluminescence, and cytokines were determined by ELISA. We found that these MC increased interleukin-8 (IL-8), cytokine-induced neutrophil chemoattractant-2alphabeta (CINC-2alphabeta) and extracellular ROS levels in human and rat neutrophils. Apart from neutrophil presence during the inflammatory process of MC-induced injury, our results suggest that hepatic neutrophil accumulation is further increased by MC-induced neutrophil-derived chemokine.
Collapse
|
174
|
Ramaiah SK, Jaeschke H. Role of neutrophils in the pathogenesis of acute inflammatory liver injury. Toxicol Pathol 2008; 35:757-66. [PMID: 17943649 DOI: 10.1080/01926230701584163] [Citation(s) in RCA: 255] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Polymorphonuclear leukocytes (neutrophils) are essential in the defense against invading microorganisms, tissue trauma or any inciting inflammatory signals. Hepatic infiltration of neutrophils is an acute response to recent or ongoing liver injury, hepatic stress or unknown systemic inflammatory signals. Once neutrophils reach the liver, they can cause mild-to-severe tissue damage and consequent liver failure. For neutrophils to appear in the liver, neutrophils have to undergo systemic activation (priming) by inflammatory mediators such as cytokines, chemokines, complement factors, immune complexes, opsonized particles and other biologically active molecules, e.g., platelet activating factor. Neutrophils accumulated in the hepatic microvasculature (sinusoids and postsinusoidal venules) can extravasate (transmigrate) into the hepatic parenchyma if they receive a signal from distressed cells. Transmigration can be mediated by a chemokine gradient established towards the hepatic parenchyma and generally involves orchestration by adhesion molecules on neutrophils (beta(2) integrins) and on endothelial cells (intracellular adhesion molecules, ICAM-1). After transmigration, neutrophils adhere to distressed hepatocytes through their beta(2) integrins and ICAM-1 expressed on hepatocytes. Neutrophil contact with hepatocytes mediate oxidative killing of hepatocytes by initiation of respiratory burst and neutrophil degranulation leading to hepatocellular oncotic necrosis. Neutrophil-mediated liver injury has been demonstrated in a variety of diseases and chemical/drug toxicities. Relevant examples are discussed in this review.
Collapse
Affiliation(s)
- Shashi K Ramaiah
- Department of Pathobiology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX 77843-4467, USA.
| | | |
Collapse
|
175
|
Hamada T, Fondevila C, Busuttil RW, Coito AJ. Metalloproteinase-9 deficiency protects against hepatic ischemia/reperfusion injury. Hepatology 2008; 47:186-98. [PMID: 17880014 DOI: 10.1002/hep.21922] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
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.
Collapse
Affiliation(s)
- Takashi Hamada
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90095-7054, USA
| | | | | | | |
Collapse
|
176
|
Cepinskas G, Katada K, Bihari A, Potter RF. Carbon monoxide liberated from carbon monoxide-releasing molecule CORM-2 attenuates inflammation in the liver of septic mice. Am J Physiol Gastrointest Liver Physiol 2008; 294:G184-91. [PMID: 17991708 DOI: 10.1152/ajpgi.00348.2007] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Recent studies suggest that exogenously administered CO is beneficial for the resolution of acute inflammation. In this study, we assessed the role of CO liberated from a systemically administered tricarbonyldichlororuthenium-(II)-dimer (CORM-2) on modulation of liver inflammation during sepsis. Polymicrobial sepsis in mice was induced by cecal ligation and perforation (CLP). CORM-2 (8 mg/kg iv) was administered immediately after CLP induction, and neutrophil [polymorphonuclear leukocyte (PMN)] tissue accumulation, activation of transcription factor, NF-kappaB, and changes in adhesion molecule ICAM-1 expression (inflammation-relevant markers) were assessed in murine liver 24 h later. In addition, the effects and potential mechanisms of CORM-2-released CO in modulation of vascular endothelial cell proinflammatory responses were assessed in vitro. To this end, human umbilical vein endothelial cells (HUVEC) were stimulated with LPS (1 microg/ml) in the presence or absence of CORM-2 (10-100 microM) and production of intracellular reactive oxygen species (ROS), (DHR123 oxidation) and NO (DAF-FM nitrosation) and subsequent activation of NF-kappaB were assessed 4 h later. In parallel, expression of ICAM-1 and inducible NO synthase (iNOS) proteins along with PMN adhesion to LPS-challenged HUVEC were also assessed. Induction of CLP resulted in increased PMN accumulation, ICAM-1 expression, and activation of NF-kappaB in the liver of septic mice. These effects were significantly attenuated by systemic administration of CORM-2. In in vitro experiments, CORM-2-released CO attenuated LPS-induced production of ROS and NO, activation of NF-kappaB, increase in ICAM-1 and iNOS protein expression and PMN adhesion to LPS-stimulated HUVEC. Taken together, these findings indicate that CO released from systemically administered CORM-2 provides anti-inflammatory effects by interfering with NF-kappaB activation and subsequent downregulation of proadhesive vascular endothelial cell phenotype in the liver of septic mice.
Collapse
Affiliation(s)
- Gediminas Cepinskas
- Centre for Critical Illness Research, Lawson Health Research Institute, London, Ontario, N6A 4G4, Canada.
| | | | | | | |
Collapse
|
177
|
Viebahn CS, Yeoh GCT. What fires prometheus? The link between inflammation and regeneration following chronic liver injury. Int J Biochem Cell Biol 2007; 40:855-73. [PMID: 18207446 DOI: 10.1016/j.biocel.2007.11.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2007] [Revised: 11/20/2007] [Accepted: 11/22/2007] [Indexed: 12/13/2022]
Abstract
Liver progenitor cells (LPCs) play a major role in the regeneration process after chronic liver damage, giving rise to hepatocytes and cholangiocytes. Thus, they provide a cell-based therapeutic alternative to organ transplant, the current treatment of choice for end-stage liver disease. In recent years, much attention has focused on unravelling the cytokines and growth factors that underlie this response. Liver regeneration following acute damage is achieved by proliferation of mature hepatocytes; yet similar cytokines, most related to the inflammatory process, are implicated in both acute and chronic liver regeneration. Thus, many recent studies represent attempts to identify LPC-specific factors. This review summarises our current understanding of LPC biology with a particular focus on the liver inflammatory response being associated with the induction of LPCs in the liver. We will describe: (i) the pathways of liver regeneration following acute and chronic damage; (ii) the similarities and differences between the two pathways; (iii) the liver inflammatory environment; (iv) the unique features of liver immunology as well as (v) the interactions between liver immune cells and LPCs. Combining data from studies on the LPC-driven regeneration process with the knowledge in the field of liver immunology will improve our understanding of the LPC response and allow us to regulate these cells in vivo and in vitro for future therapeutic strategies to treat chronic liver disease.
Collapse
Affiliation(s)
- Cornelia S Viebahn
- School of Biomedical, Biomolecular and Chemical Sciences, The University of Western Australia, 35 Stirling Highway, M310, Crawley, WA 6009, Australia.
| | | |
Collapse
|
178
|
Pritchard MT, Roychowdhury S, McMullen MR, Guo L, Arteel GE, Nagy LE. Early growth response-1 contributes to galactosamine/lipopolysaccharide-induced acute liver injury in mice. Am J Physiol Gastrointest Liver Physiol 2007; 293:G1124-33. [PMID: 17916644 DOI: 10.1152/ajpgi.00325.2007] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Early growth response (Egr)-1 is a transcription factor that regulates genes involved in inflammation, innate and adaptive immunity, coagulation, and wound healing; however, little is known about the role of Egr-1 in acute liver injury. We tested the hypothesis that Egr-1 is involved in acute liver injury induced by galactosamine/lipopolysaccharide (GalN/LPS). GalN/LPS exposure biphasically increased hepatic egr-1 mRNA accumulation at 1 h and again at 4-5.5 h after treatment in wild-type mice. Within 4-5.5 h after GalN/LPS exposure, wild-type mice exhibited histological evidence of hepatocyte injury, cell death, and extensive areas of hemorrhage, as well as increased plasma alanine aminotransferase activities. In contrast, these parameters were largely attenuated in egr-1(-/-) mice. The initial expression of tumor necrosis factor-alpha, macrophage inflammatory protein-2, monocyte chemoattractant protein-1, and intercellular adhesion molecule-1 mRNA or protein was equivalent between genotypes at 1 h after GalN/LPS administration. However, at subsequent time points, hepatic expression of these genes was decreased in egr-1(-/-) compared with wild-type mice. In addition, neutrophil extravasation from hepatic sinusoids into the liver parenchyma was decreased in egr-1(-/-) compared with wild-type mice 4 h after GalN/LPS. Whereas caspase-3 activation and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling-positive nuclei were detected in wild-type mice at 4 and 5.5 h after GalN/LPS administration, respectively, these markers of apoptosis were delayed in egr-1(-/-) mice. Delayed development of apoptosis was associated with an extension of survival by 1 h in egr-1(-/-) compared with wild-type mice. These data demonstrate that Egr-1 plays an important role in acceleration of hepatic inflammation, apoptosis, and subsequent mortality in GalN/LPS-induced acute liver injury.
Collapse
Affiliation(s)
- Michele T Pritchard
- Dept. of Pathobiology, Cleveland Clinic, 9500 Euclid Ave. NE40, Cleveland, OH 44195, USA.
| | | | | | | | | | | |
Collapse
|
179
|
MacCallum NS, Quinlan GJ, Evans TW. The Role of Neutrophil-Derived Myeloperoxidase in Organ Dysfunction and Sepsis. Intensive Care Med 2007. [DOI: 10.1007/978-0-387-49518-7_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
180
|
Lang JD, Teng X, Chumley P, Crawford JH, Isbell TS, Chacko BK, Liu Y, Jhala N, Crowe DR, Smith AB, Cross RC, Frenette L, Kelley EE, Wilhite DW, Hall CR, Page GP, Fallon MB, Bynon JS, Eckhoff DE, Patel RP. Inhaled NO accelerates restoration of liver function in adults following orthotopic liver transplantation. J Clin Invest 2007; 117:2583-91. [PMID: 17717604 PMCID: PMC1950460 DOI: 10.1172/jci31892] [Citation(s) in RCA: 177] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2007] [Accepted: 06/12/2007] [Indexed: 12/13/2022] Open
Abstract
Ischemia/reperfusion (IR) injury in transplanted livers contributes to organ dysfunction and failure and is characterized in part by loss of NO bioavailability. Inhalation of NO is nontoxic and at high concentrations (80 ppm) inhibits IR injury in extrapulmonary tissues. In this prospective, blinded, placebo-controlled study, we evaluated the hypothesis that administration of inhaled NO (iNO; 80 ppm) to patients undergoing orthotopic liver transplantation inhibits hepatic IR injury, resulting in improved liver function. Patients were randomized to receive either placebo or iNO (n = 10 per group) during the operative period only. When results were adjusted for cold ischemia time and sex, iNO significantly decreased hospital length of stay, and evaluation of serum transaminases (alanine transaminase, aspartate aminotransferase) and coagulation times (prothrombin time, partial thromboplastin time) indicated that iNO improved the rate at which liver function was restored after transplantation. iNO did not significantly affect changes in inflammatory markers in liver tissue 1 hour after reperfusion but significantly lowered hepatocyte apoptosis. Evaluation of circulating NO metabolites indicated that the most likely candidate transducer of extrapulmonary effects of iNO was nitrite. In summary, this study supports the clinical use of iNO as an extrapulmonary therapeutic to improve organ function following transplantation.
Collapse
Affiliation(s)
- John D. Lang
- Department of Anesthesiology, University of Washington School of Medicine, Seattle, Washington, USA.
Department of Pathology and
Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
Department of Biostatistics,
Department of Medicine,
Department of Surgery, and
Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Xinjun Teng
- Department of Anesthesiology, University of Washington School of Medicine, Seattle, Washington, USA.
Department of Pathology and
Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
Department of Biostatistics,
Department of Medicine,
Department of Surgery, and
Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Phillip Chumley
- Department of Anesthesiology, University of Washington School of Medicine, Seattle, Washington, USA.
Department of Pathology and
Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
Department of Biostatistics,
Department of Medicine,
Department of Surgery, and
Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jack H. Crawford
- Department of Anesthesiology, University of Washington School of Medicine, Seattle, Washington, USA.
Department of Pathology and
Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
Department of Biostatistics,
Department of Medicine,
Department of Surgery, and
Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - T. Scott Isbell
- Department of Anesthesiology, University of Washington School of Medicine, Seattle, Washington, USA.
Department of Pathology and
Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
Department of Biostatistics,
Department of Medicine,
Department of Surgery, and
Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Balu K. Chacko
- Department of Anesthesiology, University of Washington School of Medicine, Seattle, Washington, USA.
Department of Pathology and
Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
Department of Biostatistics,
Department of Medicine,
Department of Surgery, and
Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yuliang Liu
- Department of Anesthesiology, University of Washington School of Medicine, Seattle, Washington, USA.
Department of Pathology and
Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
Department of Biostatistics,
Department of Medicine,
Department of Surgery, and
Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Nirag Jhala
- Department of Anesthesiology, University of Washington School of Medicine, Seattle, Washington, USA.
Department of Pathology and
Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
Department of Biostatistics,
Department of Medicine,
Department of Surgery, and
Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - D. Ralph Crowe
- Department of Anesthesiology, University of Washington School of Medicine, Seattle, Washington, USA.
Department of Pathology and
Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
Department of Biostatistics,
Department of Medicine,
Department of Surgery, and
Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Alvin B. Smith
- Department of Anesthesiology, University of Washington School of Medicine, Seattle, Washington, USA.
Department of Pathology and
Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
Department of Biostatistics,
Department of Medicine,
Department of Surgery, and
Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Richard C. Cross
- Department of Anesthesiology, University of Washington School of Medicine, Seattle, Washington, USA.
Department of Pathology and
Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
Department of Biostatistics,
Department of Medicine,
Department of Surgery, and
Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Luc Frenette
- Department of Anesthesiology, University of Washington School of Medicine, Seattle, Washington, USA.
Department of Pathology and
Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
Department of Biostatistics,
Department of Medicine,
Department of Surgery, and
Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Eric E. Kelley
- Department of Anesthesiology, University of Washington School of Medicine, Seattle, Washington, USA.
Department of Pathology and
Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
Department of Biostatistics,
Department of Medicine,
Department of Surgery, and
Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Diana W. Wilhite
- Department of Anesthesiology, University of Washington School of Medicine, Seattle, Washington, USA.
Department of Pathology and
Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
Department of Biostatistics,
Department of Medicine,
Department of Surgery, and
Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Cheryl R. Hall
- Department of Anesthesiology, University of Washington School of Medicine, Seattle, Washington, USA.
Department of Pathology and
Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
Department of Biostatistics,
Department of Medicine,
Department of Surgery, and
Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Grier P. Page
- Department of Anesthesiology, University of Washington School of Medicine, Seattle, Washington, USA.
Department of Pathology and
Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
Department of Biostatistics,
Department of Medicine,
Department of Surgery, and
Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Michael B. Fallon
- Department of Anesthesiology, University of Washington School of Medicine, Seattle, Washington, USA.
Department of Pathology and
Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
Department of Biostatistics,
Department of Medicine,
Department of Surgery, and
Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - J. Steven Bynon
- Department of Anesthesiology, University of Washington School of Medicine, Seattle, Washington, USA.
Department of Pathology and
Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
Department of Biostatistics,
Department of Medicine,
Department of Surgery, and
Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Devin E. Eckhoff
- Department of Anesthesiology, University of Washington School of Medicine, Seattle, Washington, USA.
Department of Pathology and
Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
Department of Biostatistics,
Department of Medicine,
Department of Surgery, and
Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rakesh P. Patel
- Department of Anesthesiology, University of Washington School of Medicine, Seattle, Washington, USA.
Department of Pathology and
Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
Department of Biostatistics,
Department of Medicine,
Department of Surgery, and
Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
181
|
Ramaiah SK, Rittling S. Role of osteopontin in regulating hepatic inflammatory responses and toxic liver injury. Expert Opin Drug Metab Toxicol 2007. [DOI: 10.1517/17425255.3.4.519] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
182
|
Mookerjee RP, Stadlbauer V, Lidder S, Wright GAK, Hodges SJ, Davies NA, Jalan R. Neutrophil dysfunction in alcoholic hepatitis superimposed on cirrhosis is reversible and predicts the outcome. Hepatology 2007; 46:831-40. [PMID: 17680644 DOI: 10.1002/hep.21737] [Citation(s) in RCA: 257] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
UNLABELLED Mortality in patients with alcoholic hepatitis (AH) remains high, and although corticosteroids are widely used for treatment, the results vary considerably. In AH, neutrophils are primed and infiltrate the liver to produce injury, but paradoxically, the main cause of death in such patients is infection. Our prospective study addressed this paradox of primed neutrophils on the one hand and increased risk of infection on the other. We hypothesized that the full activation of neutrophils by a humoral factor such as endotoxin renders them unable to respond to further bacterial challenge. We analyzed neutrophil oxidative burst and phagocytosis in whole blood by fluorescence-activated cell sorting analysis in 63 alcoholic patients with cirrhosis and patients with cirrhosis with superimposed AH (cirrhosis+AH). In 16 patients, ex vivo studies determined whether the removal of endotoxin restored neutrophil function. A resting burst greater than or equal to 55[corrected]%, indicating neutrophil activation and a reduced phagocytic capacity lower than 42%, was associated with significantly greater risk of infection, organ failure, and mortality. This defective neutrophil function was transmissible through patients' plasma to normal neutrophils, and patients' neutrophil function could be restored by normal plasma. The ex vivo removal of endotoxin from patients' plasma decreased the resting burst and increased the phagocytic function. CONCLUSIONS Our study provides the rationale for a goal-directed approach to the management of patients with cirrhosis and AH, in which the assessment of neutrophil function may be an important biomarker to select patients for immunosuppressive therapy. The neutrophil dysfunction in cirrhosis and AH is reversible, with endotoxin-removal strategies providing new targets for intervention.
Collapse
Affiliation(s)
- Rajeshwar P Mookerjee
- Liver Failure Group, Institute of Hepatology, Division of Medicine, University College London, London, UK
| | | | | | | | | | | | | |
Collapse
|
183
|
Işeri SO, Sener G, Saglam B, Ercan F, Gedik N, Yeğen BC. Ghrelin alleviates biliary obstruction-induced chronic hepatic injury in rats. ACTA ACUST UNITED AC 2007; 146:73-9. [PMID: 17884193 DOI: 10.1016/j.regpep.2007.08.014] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2007] [Revised: 07/25/2007] [Accepted: 08/09/2007] [Indexed: 12/31/2022]
Abstract
BACKGROUND Reactive oxygen species and oxidative stress are implicated in hepatic stellate cell activation and liver fibrosis, which are initiated by recruitment of inflammatory cells and by activation of cytokines. OBJECTIVE The possible anti-oxidant and anti-inflammatory effects of ghrelin were evaluated in a hepatic fibrosis model in rats with bile duct ligation (BDL). METHODS Under anesthesia, bile ducts of Sprague Dawley rats were ligated, and half of the rats were subcutaneously administered with ghrelin (10 ng/kg/day) and the rest with saline for 28 days. Sham-operated control groups were administered saline or ghrelin. On the 28th day of the study, rats were decapitated and malondialdehyde (MDA) content--an index of lipid peroxidation, and myeloperoxidase (MPO) activity--an index of neutrophil infiltration--were determined in the liver tissues. Oxidant-induced tissue fibrosis was determined by collagen contents, while the hepatic injury was analyzed microscopically. Serum aspartate aminotransferase (AST), alanine aminotransferase (ALT) levels and lactate dehydrogenase (LDH) levels were determined to assess liver function and tissue damage, respectively. Pro-inflammatory cytokines; TNF-alpha, IL-1beta and IL-6 were also assayed in plasma samples. RESULTS In the saline-treated BDL group, hepatic MDA levels, MPO activity and collagen content were increased (p<0.001), suggesting oxidative organ damage, as confirmed histologically. In the ghrelin-treated BDL group, however, all of the oxidant responses were reversed significantly (p<0.05-p<0.001). Serum AST, ALT, LDH levels, and cytokines were elevated in the BDL group as compared to the control group, while this increase was significantly decreased by ghrelin treatment. CONCLUSION Owing to the anti-inflammatory and anti-oxidant effect as demonstrated in our study, it is possible to speculate that exogenously administered ghrelin may possess an antifibrotic effect against biliary obstruction-induced liver fibrosis. Thus, it seems likely that ghrelin may be of potential therapeutic value in protecting the liver fibrosis and oxidative injury due to biliary obstruction.
Collapse
Affiliation(s)
- Sevgin Ozlem Işeri
- Marmara University, School of Medicine, Department of Physiology, Haydarpaşa, Istanbul 34668, Turkey
| | | | | | | | | | | |
Collapse
|
184
|
Le Minh K, Klemm K, Abshagen K, Eipel C, Menger MD, Vollmar B. Attenuation of inflammation and apoptosis by pre- and posttreatment of darbepoetin-alpha in acute liver failure of mice. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 170:1954-63. [PMID: 17525263 PMCID: PMC1899440 DOI: 10.2353/ajpath.2007.061056] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In many liver disorders inflammation and apoptosis are important pathogenic components, finally leading to acute liver failure. Erythropoietin and its analogues are known to affect the interaction between apoptosis and inflammation in brain, kidney, and myocardium. The present study aimed to determine whether these pleiotropic actions also exert hepatoprotection in a model of acute liver injury. C57BL/6J mice were challenged with d-galactosamine (Gal) and Escherichia coli lipopolysaccharide (LPS) and studied 6 hours thereafter. Animals were either pretreated (24 hours before Gal-LPS exposure) or posttreated (30 minutes after Gal-LPS exposure) with darbepoetin-alpha (DPO, 10 mug/kg i.v.). Control mice received physiological saline. Administration of Gal-LPS caused systemic cytokine release and provoked marked hepatic damage, characterized by leukocyte recruitment and microvascular perfusion failure, caspase-3 activation, and hepatocellular apoptosis as well as enzyme release and necrotic cell death. DPO-pretreated and -posttreated mice showed diminished systemic cytokine concentrations, intrahepatic leukocyte accumulation, and hepatic perfusion failure. Hepatocellular apoptosis was significantly reduced by 50 to 75% after DPO pretreatment as well as posttreatment. In addition, treatment with DPO also significantly abrogated necrotic cell death and liver enzyme release. In conclusion, these observations may stimulate the evaluation of DPO as hepatoprotective therapy in patients with acute liver injury.
Collapse
Affiliation(s)
- Khoi Le Minh
- Institute for Experimental Surgery, University of Rostock, D-18055 Rostock, Germany
| | | | | | | | | | | |
Collapse
|
185
|
Kambayashi Y, Takekoshi S, Tanino Y, Watanabe K, Nakano M, Hitomi Y, Takigawa T, Ogino K, Yamamoto Y. Various Molecular Species of Diacylglycerol Hydroperoxide Activate Human Neutrophils via PKC Activation. J Clin Biochem Nutr 2007; 41:68-75. [PMID: 18392102 PMCID: PMC2274990 DOI: 10.3164/jcbn.2007009] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2006] [Accepted: 12/22/2006] [Indexed: 11/22/2022] Open
Abstract
We have proposed that diacylglycerol hydroperoxide-induced unregulated signal transduction causes oxidative stress-related diseases. In this study, we investigated which molecular species of diacylglycerol hydroperoxide activated human peripheral neutrophils. All diacylglycerol hydroperoxides, diacylglycerol hydroxides, and diacyglycerols tested in the present study induced superoxide production by neutrophils. The ability to activate neutrophils among molecular species containing the same fatty acid composition was as follows; diacylglycerol hydroperoxide>diacylglycerol hydroxide>/=diacylglycerol. The diacylglycerol hydroperoxide composed of linoleate was a stronger activator for neutrophils than that composed of arachidonate. 1-Palmitoyl-2-linoleoylglycerol hydroperoxide (PLG-OOH) was the strongest stimulator for neutrophils. We reconfirmed that PLG-OOH activated protein kinase C (PKC) in neutrophils. PLG-OOH induced the phosphorylation of p47(phox), a substrate of PKC and a cytosolic component of NADPH oxidase, in neutrophils, as did N-formyl-methionyl-leucyl-phenylalanine or 4beta-phorbol-12beta-myristate-13alpha-acetate. Moreover, the time course of p47(phox) phosphorylation was comparable to that of superoxide production. These results suggest that PLG-OOH activated intracellular protein kinase C. PLG-OOH, produced via an uncontrolled process, can act as a biological second messenger to cause inflammatory disease from oxidative stress.
Collapse
Affiliation(s)
- Yasuhiro Kambayashi
- Department of Environmental and Preventive Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8640, Japan
- Department of Photon and Free Radical Research¶, Japan Immunoresearch Laboratories, 351-1 Nishiyokote-cho, Takasaki 370-0021, Japan
| | - Susumu Takekoshi
- Department of Pathology, Tokai University School of Medicine, Bosedai, Isehara, Kanagawa 259-1193, Japan
| | - Yutaka Tanino
- School of Bionics, Tokyo University of Technology, 1404-1 Katakura, Hachioji 192-0982, Japan
| | - Keiichi Watanabe
- Department of Pathology, Tokai University School of Medicine, Bosedai, Isehara, Kanagawa 259-1193, Japan
| | - Minoru Nakano
- Department of Photon and Free Radical Research¶, Japan Immunoresearch Laboratories, 351-1 Nishiyokote-cho, Takasaki 370-0021, Japan
| | - Yoshiaki Hitomi
- Department of Environmental and Preventive Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8640, Japan
| | - Tomoko Takigawa
- Department of Public Health, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | - Keiki Ogino
- Department of Environmental and Preventive Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8640, Japan
- Department of Public Health, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | - Yorihiro Yamamoto
- School of Bionics, Tokyo University of Technology, 1404-1 Katakura, Hachioji 192-0982, Japan
| |
Collapse
|
186
|
Monson KM, Dowlatshahi S, Crockett ET. CXC-chemokine regulation and neutrophil trafficking in hepatic ischemia-reperfusion injury in P-selectin/ICAM-1 deficient mice. J Inflamm (Lond) 2007; 4:11. [PMID: 17524141 PMCID: PMC1891280 DOI: 10.1186/1476-9255-4-11] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2007] [Accepted: 05/24/2007] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Neutrophil adhesion and migration are critical in hepatic ischemia and reperfusion injury (I/R). P-selectin and the intercellular adhesion molecule (ICAM)-1 can mediate neutrophil-endothelial cell interactions, neutrophil migration, and the interactions of neutrophils with hepatocytes in the liver. Despite very strong preclinical data, recent clinical trials failed to show a protective effect of anti-adhesion therapy in reperfusion injury, indicating that the length of injury might be a critical factor in neutrophil infiltration. Therefore, the aim of this study was to assess the role of P-selectin and ICAM-1 in neutrophil infiltration and liver injury during early and late phases of liver I/R. METHODS Adult male wild-type and P-selectin/ICAM-1-deficient (P/I null) mice underwent 90 minutes of partial liver ischemia followed by various periods of reperfusion (6, 15 h, and a survival study). Liver injury was assessed by plasma level of alanine aminotransferase (ALT) and histopathology. The plasma cytokines, TNF-alpha, IL-6, MIP-2 and KC, were measured by ELISA. RESULTS Reperfusion caused significant hepatocellular injury in both wild-type and P/I null mice as was determined by plasma ALT levels and liver histopathology. The injury was associated with a marked neutrophil infiltration into the ischemic livers of both wild-type and P/I null mice. Although the levels of ALT and neutrophil infiltration were slightly lower in the P/I null mice compared with the wild-type mice the differences were not statistically significant. The plasma cytokine data of TNF-alpha and IL-6 followed a similar pattern to ALT data, and no significant difference was found between the wild-type and P/I null groups. In contrast, a significant difference in KC and MIP-2 chemokine levels was observed between the wild-type and P/I null mice. Additionally, the survival study showed a trend towards increased survival in the P/I null group. CONCLUSION While ICAM-1 and P-selectin does not appear to be critical for neutrophil infiltration and I/R injury in the liver, they may regulate CXC-chemokine production. Blockage of these adhesion molecules may improve survival and remote organ injury that often accompanies liver I/R injury, through chemokine regulation.
Collapse
Affiliation(s)
- Keith M Monson
- Department of Physiology, College of Human Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Shadi Dowlatshahi
- Department of Physiology, College of Human Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Elahé T Crockett
- Department of Physiology, College of Human Medicine, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
187
|
Menger MD, Vollmar B. Pathomechanisms of Ischemia-Reperfusion Injury as the Basis for Novel Preventive Strategies: Is It Time for the Introduction of Pleiotropic Compounds? Transplant Proc 2007; 39:485-8. [PMID: 17362764 DOI: 10.1016/j.transproceed.2007.01.022] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Ischemia-reperfusion-associated tissue dysfunction and organ failure still represent major complications in transplantation surgery. The pathomechanisms involve microvascular perfusion failure, ie, no-reflow and tissue hypoxia despite reperfusion and reoxygenation. However, postischemic reperfusion also provokes an inflammatory response, ie, reflow paradox, with activation of macrophages, recruitment of leukocytes, and accumulation of platelets, involving surface adhesion molecules such as P-selectin, P-selectin glycoprotein ligand (PSGL)-1, Mac-1, and intercellular adhesion molecule (ICAM)-1. These inflammatory cells produce cytokines, chemokines, lipid mediators, and oxygen radicals, which all may contribute to the manifestation of injury, including apoptosis, necrosis, and necrapoptosis. Although specific inhibition of single mediators, such as tumor necrosis factor (TNF)-alpha, interleukin (IL)-1, and oxygen radicals, or distinct molecules, such as P-selectin and ICAM-1, has been shown to be protective in the experimental setting, these single-agent antimediator and antimolecule approaches did not find their way into clinical practice. Clinically, University of Wisconsin (UW) solution for organ preservation is still the major milestone for prevention of ischemia- and reperfusion-associated injury. Characteristically, this treatment strategy does not represent an anti-single mediator approach, but exerts protection by influencing multiple pathways involved in hypoxic and inflammatory injury, potentially restoring the overall homeostasis. This type of pleiotropic action may also be achieved by single pharmacological compounds, such as statins, erythropoietin, hemoxygenase-1, and L-glycine. In recent experimental studies, these compounds have been shown to be effective to reduce post-ischemic-reperfusion injury, and, additionally, to be associated with less side effects. Accordingly, these pleiotropic substances may represent ideal candidates for pharmacological preconditioning in patient treatment, and, thus, should be further evaluated in clinical trials.
Collapse
Affiliation(s)
- M D Menger
- Institute for Clinical & Experimental Surgery, University of Saarland, Homburg/Saar, Germany
| | | |
Collapse
|
188
|
MacCallum NS, Quinlan GJ, Evans TW. The Role of Neutrophil-Derived Myeloperoxidase in Organ Dysfunction and Sepsis. YEARBOOK OF INTENSIVE CARE AND EMERGENCY MEDICINE 2007. [DOI: 10.1007/978-3-540-49433-1_16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|