1
|
Cheng YJ, Shyu WC, Teng YH, Lan YH, Lee SD. Antagonistic interaction between cordyceps sinensis and exercise on protection in fulminant hepatic failure. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2015; 42:1199-213. [PMID: 25242080 DOI: 10.1142/s0192415x1450075x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Herb supplements are widely used by Asian athletes; however, there are no studies evaluated the co-effects of exercise and herb supplements on hepatic failure. In this study, D-GalN/LPS-induced fulminant hepatic failure was used to examine whether there are synergistic or antagonistic effects of exercise and Cordyceps sinensis (CS). Mice were randomly divided into eight groups: control, swimming exercise for four weeks, D-GalN/LPS challenge, swimming exercise plus D-GalN/LPS, 20 mg/kg or 40 mg/kg CS pretreated for four weeks plus D-GalN/LPS, and swimming exercise combined with 20 mg/kg or 40 mg/kg CS pretreatment plus D-GalN/LPS. Either exercise or 40 mg/kg CS pretreatment alone significantly decreased D-GalN/LPS-induced TNF-α, AST, NO, apoptotic-related proteins, and hepatocyte apoptosis. Exercise or 40 mg/kg CS alone increased the IL-10 and D-GalN/LPS-suppressed Superoxide Dismutase (SOD) level. However, no protective or worse effect was observed in the mice treated with exercise preconditioning combined 40 mg/kg CS compared to those receive exercise alone or CS alone. TNF-α, AST, NO level, caspase-3 activity, and hepatocytes apoptosis were not significantly different in the exercise combined with 40 mg/kg CS compared to mice challenged with D-GalN/LPS. The IL-10 level was significantly decreased after D-GalN/LPS stimulation in the mice received exercise combined with 40 mg/kg CS, indicating the combination strongly reduced the anti-inflammatory effect. In summary, preconditioning exercise or CS pretreatment alone can protect mice from septic liver damage, but in contrast, the combination of exercise and CS does not produce any benefit. The antagonistic interactions between exercise and CS imply taking CS is not recommended for people who undertake regular exercise.
Collapse
Affiliation(s)
- Yu-Jung Cheng
- Department of Physical Therapy and Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan, ROC
| | | | | | | | | |
Collapse
|
2
|
Shinozaki S, Chang K, Sakai M, Shimizu N, Yamada M, Tanaka T, Nakazawa H, Ichinose F, Yamada Y, Ishigami A, Ito H, Ouchi Y, Starr ME, Saito H, Shimokado K, Stamler JS, Kaneki M. Inflammatory stimuli induce inhibitory S-nitrosylation of the deacetylase SIRT1 to increase acetylation and activation of p53 and p65. Sci Signal 2014; 7:ra106. [PMID: 25389371 DOI: 10.1126/scisignal.2005375] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Inflammation increases the abundance of inducible nitric oxide synthase (iNOS), leading to enhanced production of nitric oxide (NO), which can modify proteins by S-nitrosylation. Enhanced NO production increases the activities of the transcription factors p53 and nuclear factor κB (NF-κB) in several models of disease-associated inflammation. S-nitrosylation inhibits the activity of the protein deacetylase SIRT1. SIRT1 limits apoptosis and inflammation by deacetylating p53 and p65 (also known as RelA), a subunit of NF-κB. We showed in multiple cultured mammalian cell lines that NO donors or inflammatory stimuli induced S-nitrosylation of SIRT1 within CXXC motifs, which inhibited SIRT1 by disrupting its ability to bind zinc. Inhibition of SIRT1 reduced deacetylation and promoted activation of p53 and p65, leading to apoptosis and increased expression of proinflammatory genes. In rodent models of systemic inflammation, Parkinson's disease, or aging-related muscular atrophy, S-nitrosylation of SIRT1 correlated with increased acetylation of p53 and p65 and activation of p53 and NF-κB target genes, suggesting that S-nitrosylation of SIRT1 may represent a proinflammatory switch common to many diseases and aging.
Collapse
Affiliation(s)
- Shohei Shinozaki
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, MA 02129, USA. Department of Geriatrics and Vascular Medicine, Tokyo Medical and Dental University Graduate School, Tokyo 113-8519, Japan
| | - Kyungho Chang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, MA 02129, USA. Department of Anesthesiology and Pain Relief Center, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Michihiro Sakai
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, MA 02129, USA
| | - Nobuyuki Shimizu
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, MA 02129, USA
| | - Marina Yamada
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, MA 02129, USA
| | - Tomokazu Tanaka
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, MA 02129, USA
| | - Harumasa Nakazawa
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, MA 02129, USA
| | - Fumito Ichinose
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, MA 02129, USA
| | - Yoshitsugu Yamada
- Department of Anesthesiology and Pain Relief Center, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Akihito Ishigami
- Tokyo Metropolitan Institute of Gerontology, Tokyo 173-0015, Japan
| | - Hideki Ito
- Tokyo Metropolitan Institute of Gerontology, Tokyo 173-0015, Japan
| | - Yasuyoshi Ouchi
- Department of Geriatric Medicine, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan. Federation of National Public Service Personnel Mutual Aid Associations Toranomon Hospital, Tokyo 105-0001, Japan
| | - Marlene E Starr
- Department of Surgery, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Hiroshi Saito
- Department of Surgery, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Kentaro Shimokado
- Department of Geriatrics and Vascular Medicine, Tokyo Medical and Dental University Graduate School, Tokyo 113-8519, Japan
| | - Jonathan S Stamler
- Institute for Transformative Molecular Medicine and Harrington Discovery Institute, Case Western Reserve University and University Hospital, Cleveland, OH 44106, USA
| | - Masao Kaneki
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, MA 02129, USA.
| |
Collapse
|
3
|
Fukazawa K, Pretto EA. The effect of methylene blue during orthotopic liver transplantation on post reperfusion syndrome and postoperative graft function. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2011; 18:406-13. [PMID: 21104279 DOI: 10.1007/s00534-010-0344-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND/PURPOSE In orthotopic liver transplantation (OLT), a major component of the post-reperfusion syndrome is hypotension, which may lead to additional graft liver ischemia-reperfusion injury. A proposed mechanism of reperfusion hypotension is the massive induction of oxidative stress triggering the release of pro-inflammatory mediators, including nitric oxide (NO). Methylene blue (MB) is an inhibitor of inducible NO synthase and an NO scavenger that has been shown to attenuate reperfusion hypotension. Of note, recent reports have shown that the exogenous administration of NO during OLT significantly improved the recovery of the graft liver. Therefore, we sought to investigate the effects of MB on the functional recovery of the graft liver following OLT. METHODS We analyzed retrospective data from 715 patients who underwent OLT between 2003 and 2008. We classified patients into those who received a 1-1.5 mg/kg intravenous bolus of MB immediately prior to reperfusion (MB group) and those who did not (control group). Propensity score matching was used to adjust for differences between patients who received intraoperative MB and those who did not, and these data were used to determine the association between a single MB bolus during OLT and postoperative graft dysfunction. RESULTS Our study cohort consisted of 715 OLT patients, of whom 105 received MB and 610 did not. After propensity score matching, demographic and donor data were similar in the two groups, except for the older age of recipients in the MB group (55.5 ± 0.9 vs 53.1 ± 0.8 years, p = 0.026). No differences were seen in mean arterial pressure changes after reperfusion and no differences were found in vasopressor requirements (bolus or infusion) or transfusion requirements. In addition, there was no significant difference in the incidence of primary nonfunction, retransplantation within 60 days, acute rejection, or graft survival between the groups by multivariate analysis or Kaplan-Meier survival analysis. CONCLUSIONS In our study, the administration of MB at 1-1.5 mg/kg immediately prior to reperfusion did not prevent post-reperfusion hypotension and did not decrease vasopressor usage or transfusion requirements after reperfusion. Also, MB did not have any impact on postoperative graft function. These findings may argue against the routine use of MB during OLT.
Collapse
Affiliation(s)
- Kyota Fukazawa
- Division of Solid Organ Transplantation, Department of Anesthesiology, Preoperative and Pain Management, Leonard Miller School of Medicine, University of Miami, 1611 NW 12th Avenue, D318, Miami, FL 33136, USA.
| | | |
Collapse
|
4
|
Suliburk JW, Ward JL, Helmer KS, Adams SD, Zuckerbraun BS, Mercer DW. Ketamine-induced hepatoprotection: the role of heme oxygenase-1. Am J Physiol Gastrointest Liver Physiol 2009; 296:G1360-9. [PMID: 19372106 PMCID: PMC2697945 DOI: 10.1152/ajpgi.00038.2009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Lipopolysaccharide (LPS) causes hepatic injury that is mediated, in part, by upregulation of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). Ketamine has been shown to prevent these effects. Because upregulation of heme oxygenase-1 (HO-1) has hepatoprotective effects, as does carbon monoxide (CO), an end product of the HO-1 catalytic reaction, we examined the effects of HO-1 inhibition on ketamine-induced hepatoprotection and assessed whether CO could attenuate LPS-induced hepatic injury. One group of rats received ketamine (70 mg/kg ip) or saline concurrently with either the HO-1 inhibitor tin protoporphyrin IX (50 micromol/kg ip) or saline. Another group of rats received inhalational CO (250 ppm over 1 h) or room air. All rats were given LPS (20 mg/kg ip) or saline 1 h later and euthanized 5 h after LPS or saline. Liver was collected for iNOS, COX-2, and HO-1 (Western blot), NF-kappaB and PPAR-gamma analysis (EMSA), and iNOS and COX-2 mRNA analysis (RT-PCR). Serum was collected to measure alanine aminotransferase as an index of hepatocellular injury. HO-1 inhibition attenuated ketamine-induced hepatoprotection and downregulation of iNOS and COX-2 protein. CO prevented LPS-induced hepatic injury and upregulation of iNOS and COX-2 proteins. Although CO abolished the ability of LPS to diminish PPAR-gamma activity, it enhanced NF-kappaB activity. These data suggest that the hepatoprotective effects of ketamine are mediated primarily by HO-1 and its end product CO.
Collapse
Affiliation(s)
- James W. Suliburk
- Department of Surgery, The University of Texas Medical School at Houston, Houston, Texas; and Department of Surgery, The University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jeremy L. Ward
- Department of Surgery, The University of Texas Medical School at Houston, Houston, Texas; and Department of Surgery, The University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Kenneth S. Helmer
- Department of Surgery, The University of Texas Medical School at Houston, Houston, Texas; and Department of Surgery, The University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Sasha D. Adams
- Department of Surgery, The University of Texas Medical School at Houston, Houston, Texas; and Department of Surgery, The University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Brian S. Zuckerbraun
- Department of Surgery, The University of Texas Medical School at Houston, Houston, Texas; and Department of Surgery, The University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - David W. Mercer
- Department of Surgery, The University of Texas Medical School at Houston, Houston, Texas; and Department of Surgery, The University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
5
|
Pretreatment with bone morphogenetic protein-7 (BMP-7) mimics ischemia preconditioning following intestinal ischemia/reperfusion injury in the intestine and liver. Shock 2009; 30:532-6. [PMID: 18461025 DOI: 10.1097/shk.0b013e31816f20f1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Intestinal ischemia/reperfusion (I/R) injury has been shown to cause intestinal mucosal injury and adversely affect function. Ischemic preconditioning (IPC) has been shown to protect against intestinal I/R injury by reducing polymorphonuclear leukocyte infiltration, intestinal mucosal injury, and liver injury, and preserve intestinal transit. Bone morphogenetic protein 7 (BMP-7) has been shown to protect against I/R injury in the kidney and brain. Recently, microarray analysis has been used to examine the possible IPC candidate pathways. This work revealed that IPC may work through upregulation of BMP-7. The purpose of this study was to examine if pretreatment with BMP-7 would replicate the effects seen with IPC in the intestine and liver after intestinal I/R. Rats were randomized to six groups: sham, I/R (30 min of superior mesenteric artery occlusion and 6 h of R), IPC+R (three cycles of superior mesenteric artery occlusion for 4 min and R for 10 min), IPC+I/R, BMP-7+R (100 microm/kg recombinant human BMP-7), or BMP-7+I/R. A duodenal catheter was placed, and 30 min before sacrifice, fluorescein isothiocyanate-Dextran was injected. At sacrifice, dye concentrations were measured to determine intestinal transit. Ileal mucosal injury was determined by histology and myeloperoxidase activity was used as a marker of polymorphonuclear leukocyte infiltration. Serum levels of aspartate aminotransferase were measured at sacrifice to determine liver injury. Pretreatment with BMP-7 significantly improved intestinal transit and significantly decreased intestinal mucosal injury and serum aspartate aminotransferase levels, comparable to animals undergoing IPC. In conclusion, BMP-7 protected against intestinal I/R-induced intestinal and liver injury. Bone morphogenetic protein 7 may be a more logical surrogate to IPC in the prevention of injury in the setting of intestinal I/R.
Collapse
|
6
|
Fasting exacerbates and feeding diminishes LPS-induced liver injury in the rat. Dig Dis Sci 2009; 54:767-73. [PMID: 18688715 DOI: 10.1007/s10620-008-0425-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2008] [Accepted: 06/25/2008] [Indexed: 12/09/2022]
Abstract
INTRODUCTION Enteral nutrition improves clinical outcomes. The effects of feeding on LPS induced liver injury are unknown. We hypothesized that feeding would attenuate liver injury from LPS. METHODS Fasted or fed rats were given LPS (20 mg/kg i.p.) or saline for 5 h and sacrificed. Serum aminotransferases and cytokines (immunoassay) were measured. Oxidative stress protein (iNOS, COX2, and HO1) assessments (Western immunoblot) were also obtained. RESULTS In fasted rats, LPS significantly increased serum aminotransferase levels, enhanced hepatic COX2, iNOS, and HO1 immunoreactivity, and increased serum cytokine levels when compared to controls. While feeding diminished liver enzymes, attenuated expression of COX2 and iNOS, and blunted production of pro-inflammatory cytokines, it did not modulate LPS-induced expression of the anti-inflammatory markers HO1 and IL-10. CONCLUSION These data suggest that feeding decreases liver injury by attenuating expression of pro-inflammatory mediators while maintaining expression of anti-inflammatory mediators, both systemically and locally.
Collapse
|
7
|
Tamandl D, Jørgensen P, Gundersen Y, Fuegger R, Sautner T, Aasen AO, Goetzinger P. Nitric oxide administration restores the hepatic artery buffer response during porcine endotoxemia. J INVEST SURG 2008; 21:183-94. [PMID: 18615315 DOI: 10.1080/08941930802130131] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The hepatic artery buffer response, which is lost during endotoxemia, plays a central role in the autoregulation of liver perfusion. A temporarily decreased synthesis of nitric oxide during early endotoxemia might be responsible for this dysfunction; hence exogenous administration of nitric oxide could reestablish the autoregulation of hepatic blood flow and help prevent hepatic damage later in septic shock. Fifteen pigs were treated with lipopolysaccharide +/- the nitric oxide donor nitroprusside-sodium via the portal vein. Hemodynamics were measured, and serum chemistry and liver biopsies for nitric oxide synthase expression were obtained. Lipopolysaccharide decreased arterial liver perfusion after 5 hours by 38% (p = .012), which was reversed by addition of nitroprusside (8%). Administration of nitroprusside preserved an increase of 28% in hepatic arterial upon portal vein flow reduction (p < .001). Nitroprusside maintained mRNA levels of constitutive nitric oxide synthase in liver tissue which were decreased by lipopolysaccharide (p = .026 vs. p = .114) and tempered the burst in inducible nitric oxide synthase expression at t = 3 hours. The early administration of the nitric oxide donor sodium nitroprusside during endotoxemia is able to reestablish the autoregulatory response of the hepatic artery following reduction of hepatic blood flow. This beneficial effect might help to prevent subsequent hepatic damage in the course of abdominal sepsis.
Collapse
Affiliation(s)
- Dietmar Tamandl
- Department of Surgery, Vienna General Hospital, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
8
|
Liver Tissue Inducible Nitric Oxide Synthase (iNOS) Expression and Lipid Peroxidation in Experimental Hepatic Ischemia Reperfusion Injury Stimulated With Lipopolysaccharide: The Role of Aminoguanidine. J Surg Res 2008; 148:214-23. [DOI: 10.1016/j.jss.2007.10.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2007] [Revised: 09/14/2007] [Accepted: 10/12/2007] [Indexed: 11/19/2022]
|
9
|
Abstract
BACKGROUND The pathophysiology of sepsis is incompletely understood, however alterations in systemic inflammation and serum cytokines are thought to play a central role. In the rat, ketamine, but not isoflurane, prevents hepatic injury from lipopolysaccharide (LPS). The effect of these anesthetics on the systemic inflammatory response and other organs remains to be fully elucidated. We hypothesized that ketamine, but not isoflurane, would blunt the cytokine response to LPS administration. METHODS Male rats received no anesthesia, intraperitoneal ketamine (70 mg/kg), or inhalational isoflurane. One hour later, LPS (20 mg/kg, intraperitoneal) or saline was given for 5 hours and rats were killed. Gastric fluid volumes were determined as an index of gastric emptying. Serum was collected and cytokines measured via a multiplexed suspension immunoassay. RESULTS In nonanesthetized rats, LPS increased gastric luminal fluid accumulation and serum levels of proinflammatory cytokines when compared with saline controls. Anesthesia with either ketamine or isoflurane caused a significant reduction in LPS-induced changes in serum cytokines, although ketamine had a more dramatic reduction in tumor necrosis factor alpha levels than did isoflurane. Both anesthetics reduced the interleukin IL-6/IL-10 ratio in response to LPS when compared with LPS alone. Ketamine, but not isoflurane, prevented LPS-induced gastric luminal fluid accumulation. CONCLUSIONS These data indicate that both ketamine and isoflurane diminish the systemic inflammatory response to LPS in the rat as measured by serum cytokines and a reduced IL-6/IL-10 ratio. However, only ketamine improves LPS-induced gastric dysfunction, perhaps secondary to its ability to reduce serum tumor necrosis factor alpha levels more effectively.
Collapse
|
10
|
Kan WH, Hsu JT, Schwacha MG, Choudhry MA, Raju R, Bland KI, Chaudry IH. Selective inhibition of iNOS attenuates trauma-hemorrhage/resuscitation-induced hepatic injury. J Appl Physiol (1985) 2008; 105:1076-82. [PMID: 18635878 DOI: 10.1152/japplphysiol.90495.2008] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Although trauma-hemorrhage produces tissue hypoxia, systemic inflammatory response and organ dysfunction, the mechanisms responsible for these alterations are not clear. Using a potent selective inducible nitric oxide (NO) synthase inhibitor, N-[3-(aminomethyl) benzyl]acetamidine (1400W), and a nonselective NO synthase inhibitor, N(G)-nitro-L-arginine methyl ester (L-NAME), we investigated whether inducible NO synthase plays any role in producing hepatic injury, inflammation, and changes of protein expression following trauma-hemorrhage. To investigate this, male Sprague-Dawley rats were subjected to midline laparotomy and hemorrhagic shock (mean blood pressure 35-40 mmHg for approximately 90 min) followed by fluid resuscitation. Animals were treated with either vehicle (DMSO) or 1400W (10 mg/kg body wt ip), or L-NAME (30 mg/kg iv), 30 min before resuscitation and killed 2 h after resuscitation. Trauma-hemorrhage/resuscitation induced a marked hypotension and increase in markers of hepatic injury (i.e., plasma alpha-glutathione S-transferase, tissue myeloperoxidase activity, and nitrotyrosine formation). Hepatic expression of iNOS, hypoxia-inducible factor-1alpha, ICAM-1, IL-6, TNF-alpha, and neutrophil chemoattractant (cytokine-induced neutrophil chemoattractant-1 and macrophage inflammatory protein-2) protein levels were also markedly increased following trauma-hemorrhage/resuscitation. Administration of the iNOS inhibitor 1400W significantly attenuated hypotension and expression of these mediators of hepatic injury induced by trauma-hemorrhage/resuscitation. However, administration of L-NAME could not attenuate hepatic dysfunction and tissue injury mediated by trauma-hemorrhage, although it improved mean blood pressure as did 1400W. These results indicate that increased expression of iNOS following trauma-hemorrhage plays an important role in the induction of hepatic damage under such conditions.
Collapse
Affiliation(s)
- Wen-Hong Kan
- Center for Surgical Research, University of Alabama at Birmingham, 1670 University Blvd., Birmingham, AL 35294-0019, USA
| | | | | | | | | | | | | |
Collapse
|
11
|
Suliburk J, Helmer K, Moore F, Mercer D. The gut in systemic inflammatory response syndrome and sepsis. Enzyme systems fighting multiple organ failure. Eur Surg Res 2007; 40:184-9. [PMID: 17998777 DOI: 10.1159/000110859] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2007] [Accepted: 08/29/2007] [Indexed: 12/13/2022]
Abstract
The prognosis and care of critically ill ICU patients has improved over recent years, but the development of multiple organ failure (MOF) continues to cause significant morbidity and mortality. Shock, with resultant organ ischemia, appears to play a critical role in the development of MOF. It is our global hypothesis that MOF is a gut-derived phenomenon and that novel interventions can improve outcome in shock-induced gut inflammation and dysfunction in critically ill patients. We have found that the anesthetic agent ketamine has a profound impact on the response to endotoxic shock. This review summarizes our findings on the mechanisms of action by which ketamine is able to modulate the nitric oxide, cyclo-oxygenase and heme-oxygenase enzyme systems to attenuate endotoxin-induced organ dysfunction.
Collapse
Affiliation(s)
- J Suliburk
- The University of Texas Medical School, Department of Surgery, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
12
|
Cavriani G, Domingos HV, Oliveira-Filho RM, Sudo-Hayashi LS, Vargaftig BB, de Lima WT. Lymphatic thoracic duct ligation modulates the serum levels of IL-1beta and IL-10 after intestinal ischemia/reperfusion in rats with the involvement of tumor necrosis factor alpha and nitric oxide. Shock 2007; 27:209-13. [PMID: 17224798 DOI: 10.1097/01.shk.0000238068.84826.52] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Intestinal ischemia/reperfusion (I/R) causes local and remote injuries that are multifactorial and essentially inflammatory in nature. To study the putative influences of nitric oxide (NO) and tumor necrosis factor alpha (TNF-alpha) on the release of interleukin (IL) 1beta and IL-10 and the involvement of lymphatic system on a systemic inflammation caused by I/R, we have quantified the serum and lymph levels of IL-1beta and IL-10 in rats during I/R after treatment with inhibitors of NO synthase (N-nitro-L-arginine methyl ester hydrochloride [L-NAME]) or TNF-alpha (pentoxifylline [PTX]). Intestinal I/R was performed by means of a 45-min occlusion of the mesenteric artery, followed by 2-h reperfusion; groups of rats subjected to I/R had the thoracic lymph duct ligated immediately before the procedure. The I/R caused a significant increase of the serum levels of IL-1beta and IL-10 in rats with intact thoracic lymph duct, whereas the thoracic duct ligation blunted the serum release of IL-1beta and elevated that of IL-10. The levels of the cytokines collected in the lymph after I/R increased, and even more increase was observed in L-NAME-treated rats. L-NAME significantly increased the lymph levels of IL-1beta and IL-10; in serum, however, only IL-1beta increased in rats with either intact or ligated thoracic lymph duct. The treatment with PTX reduced the serum levels of IL-1beta irrespective of the lymph circulation interruption but was effective to increase the IL-10 levels in intact rats during I/R. The lymphatic levels of IL-1beta of rats subjected to I/R were reduced and those of IL-10 were increased after treatment with PTX. In conclusion, during I/R, the serum levels of IL-1beta seem modulated by stimulant mechanisms that could be associated with TNF-alpha and inhibited by NO and by the integrity of the thoracic lymphatic flow. On the other hand, IL-10 seems controlled by TNF-alpha-related, largely NO-independent mechanisms. Thus, it is reasonable to suppose that an endogenous mechanism that can limit the systemic inflammatory response ensuing an I/R splanchnic trauma exists.
Collapse
Affiliation(s)
- Gabriela Cavriani
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
13
|
Eum HA, Yeom DH, Lee SM. Role of nitric oxide in the inhibition of liver cytochrome P450 during sepsis. Nitric Oxide 2006; 15:423-31. [PMID: 16884934 DOI: 10.1016/j.niox.2006.06.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2006] [Revised: 05/18/2006] [Accepted: 06/19/2006] [Indexed: 10/24/2022]
Abstract
Nitric oxide (NO) plays an important role in the pathophysiology of sepsis and septic shock but the mechanism is not well understood. The aim of this study was to investigate the role of NO in the cytochrome P450 (CYP) isozyme activity and the expression of its gene during polymicrobial sepsis. The rats were subjected to polymicrobial sepsis by cecal ligation and puncture (CLP). Aminoguanidine (AG, 100 mg/kg body weight) or N(omega)-nitro-L-arginine methyl ester (L-NAME, 100 mg/kg body weight) was injected intraperitoneally at 0, 3, 6, 10, and 20 h after CLP. The plasma nitrite/nitrate concentration increased 24 h after CLP, and this increase was almost completely abolished by AG and L-NAME. Sepsis increased the serum aminotransferase and lipid peroxidation levels, which were attenuated by AG but augmented by L-NAME. The hepatic concentration of the reduced gluthathione decreased in the CLP rats, which was inhibited by AG but augmented by L-NAME. The total CYP content decreased after CLP, which was restored by AG and L-NAME. The CYP1A1, 1A2, and 2E1 activities, along with their protein levels, decreased 24 h after CLP but these decreases were reversed by AG and L-NAME. The CYP1A1, 1A2, 2B1, and 2E1 mRNA expression levels decreased 24 h after CLP, and L-NAME inhibited this decrease. NO plays a key role in the sepsis-mediated decrease in CYP via the interplay of two different mechanisms: NO-dependent suppression of protein via the enhanced inducible NO synthase, and NO-dependent transcriptional suppression via endothelial NO synthase.
Collapse
Affiliation(s)
- Hyun-Ae Eum
- College of Pharmacy, Sungkyunkwan University, Suwon 440-746, South Korea
| | | | | |
Collapse
|
14
|
Hara Y, Teramoto K, Ishidate K, Arii S. Cytoprotective function of tetrahydrobiopterin in rat liver ischemia/reperfusion injury. Surgery 2006; 139:377-84. [PMID: 16546503 DOI: 10.1016/j.surg.2005.08.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2005] [Revised: 08/18/2005] [Accepted: 08/19/2005] [Indexed: 11/23/2022]
Abstract
BACKGROUND Tetrahydrobiopterin (BH(4)) is a key coenzyme of nitric oxide synthase (NOS), which is associated with a cytoprotective function in various ischemia-reperfusion (I/R) injury models. There have been a few reports on the efficacy of BH(4) in the treatment of I/R injury in other organs; therefore, the aim of this study was to investigate the effect of BH(4) related with NOS reaction in hepatic I/R injury. METHODS A model of 70% liver I/R injury with a 100-minute ischemic time was created in rats, and the non-ischemic lobes were then resected. The rats were given BH(4) (BH(4) group) or saline solution (saline group) before reperfusion. The specific inducible NOS blocker 1400W was used to evaluate the effect of endogenous inducible NOS in the I/R hepatic injury. Survival, nitric oxide products (nitrate and nitrite), NOS expression, and nitrotyrosine (ie, the peroxynitrite product) were measured after reperfusion. RESULTS On day 7, the survival rate was 62.5% in the BH(4) group, as opposed to 14.3% in the saline group (P = .0004); 1400W administration to the BH(4) group decreased the survival rate to 0% (P = .003). BH(4) prevented the significant increase in total bilirubin levels (P < .01) after 12-hour reperfusion. The increases in serum alanine transaminase levels (after 3 hours and 12 hours of reperfusion) were significantly (P < .01) attenuated in the BH(4) group. BH(4) increased the nitrate/nitrite concentrations in liver tissue (P < .05) and reduced nitrotyrosine production, and the protein assay showed that BH(4) increased inducible NOS and endothelial NOS expression. Histologic examination of the liver revealed that BH(4) mitigated the damage that was caused by liver I/R. CONCLUSION Exogenous BH(4) increased nitric oxide production, which attenuated the hepatic I/R injury.
Collapse
Affiliation(s)
- Yuzuru Hara
- Department of Surgery, School of Medicine, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | | | | | | |
Collapse
|
15
|
Suliburk JW, Helmer KS, Gonzalez EA, Robinson EK, Mercer DW. Ketamine attenuates liver injury attributed to endotoxemia: role of cyclooxygenase-2. Surgery 2005; 138:134-40. [PMID: 16153419 DOI: 10.1016/j.surg.2005.03.024] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2004] [Revised: 03/23/2005] [Accepted: 03/25/2005] [Indexed: 01/12/2023]
Abstract
BACKGROUND Endotoxic shock can cause end-organ dysfunction and liver injury. Critically ill patients frequently require surgical intervention under general anesthesia for source control. However, the effects of anesthetics on organ function during sepsis and their influence on inflammatory mediators such as cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) remain to be fully elucidated. Because ketamine anesthesia has anti-inflammatory effects in some tissues, we hypothesized that it would attenuate lipopolysaccharide (LPS)-induced liver injury. METHODS Adult rats were given no anesthesia (saline), continuous isoflurane inhalation, or intraperitoneal (i.p.) injection of ketamine 70 mg/kg. One hour later, the rats received saline or LPS (20 mg/kg i.p.) for 5 hours. The rats were killed, and serum hepatocellular enzymes, liver COX-2, iNOS protein (Western immunoblot), and nuclear factor kappa B (NF-kappaB)-binding activity (electrophoretic mobility shift assay) determined. In a separate study, the role of COX-2 in LPS-induced liver injury was examined by pretreating rats with the selective COX-2 inhibitor NS-398 (3 mg/kg, i.p.) and the role of iNOS examined with the use of the selective inhibitor aminoguanidine (45 mg/kg, i.p.) 1 hour before LPS. RESULTS LPS increased serum aspartate aminotransferase and alanine aminotransferase levels, hepatic iNOS and COX-2 protein, and nuclear factor NF-kappaB. Ketamine, but not isoflurane, attenuated these effects caused by LPS. COX-2 inhibition with NS-398 as well as iNOS inhibition with aminoguanidine diminished LPS-induced changes in aspartate aminotransferase and alanine aminotransferase levels. CONCLUSIONS These data indicate that anesthetics differ in their effects on liver injury caused by LPS. Ketamine has hepatoprotective effects, while isoflurane does not. Moreover, the protective effects of ketamine are mediated, at least in part, through a reduction in COX-2 and iNOS protein that could be regulated via changes in NF-kappaB-binding activity.
Collapse
Affiliation(s)
- James W Suliburk
- Department of Surgery, The University of Texas Medical School at Houston, 77030, USA.
| | | | | | | | | |
Collapse
|
16
|
Wei CL, Hon WM, Lee KH, Khoo HE. Chronic administration of aminoguanidine reduces vascular nitric oxide production and attenuates liver damage in bile duct-ligated rats. Liver Int 2005; 25:647-56. [PMID: 15910502 DOI: 10.1111/j.1478-3231.2005.01063.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Nitric oxide (NO) has been implicated in the pathogenesis of liver cirrhosis. This study investigated the activity of nitric oxide synthase (NOS) in cirrhosis induced by bile duct-ligation (BDL) with NOS inhibitors. METHOD Three days after operation, rats were randomized to receive aminoguanidine (AG, 25 mg/kg/day) or L-N(G)-nitro-L-arginine methyl ester (L-NAME, 10 mg/kg/day) for 21 days. RESULTS Vascular NO production, which was increased in BDL cirrhotic rats, was reduced by 75% with AG but not L-NAME chronic administration. AG treatment attenuated liver damage, while L-NAME aggravated it. AG significantly suppressed inducible NOS (iNOS) expression in aorta of BDL rats at both mRNA and protein level, but much less efficient in reducing it in liver. In contrast, endothelial NOS (eNOS) expression was not markedly affected. Calcium-independent NOS activity, which was dramatically increased in aorta of BDL rats, was abolished by AG treatment. In liver, however, both calcium-dependent and -independent NOS activity were increased by AG treatment. CONCLUSION Chronic administration of AG could reduce systemic NO levels as well as suppress iNOS expression and activity in aorta of BDL rats. It also improved liver function, possibly because of its ability to increase hepatic NOS activity, and to correct the systemic hemodynamic disorders by decreasing vascular NO production.
Collapse
Affiliation(s)
- Chang-Li Wei
- Department of Paediatrics, Faculty of Medicine, National University of Singapore, 10 Kent Ridge Crescent, Singapore 119260, Singapore
| | | | | | | |
Collapse
|
17
|
Suliburk JW, Gonzalez EA, Kennison SD, Helmer KS, Mercer DW. Differential effects of anesthetics on endotoxin-induced liver injury. ACTA ACUST UNITED AC 2005; 58:711-6; discussion 716-7. [PMID: 15824646 DOI: 10.1097/01.ta.0000159245.60495.00] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND The liver is both a source and a target of inflammatory and anti-inflammatory mediators during sepsis. The oxidative stress proteins inducible nitric oxide synthase (iNOS) and heme oxygenase-1 (HO-1) are upregulated in the liver during sepsis but have opposite roles. Upregulation of HO-1 has hepatoprotective effects, whereas iNOS has injurious effects to the liver. Although recent studies indicate that ketamine anesthesia has anti-inflammatory effects during sepsis, the effects of other anesthetics are unknown. We hypothesized that ketamine, but not isoflurane, would attenuate lipopolysaccharide (LPS)-induced liver injury through differential modulation of iNOS and HO-1. METHODS Adult rats were given no anesthesia (saline), continuous isoflurane inhalation, or intraperitoneal ketamine (70 mg/kg). One hour later, saline or LPS (20 mg/kg intraperitoneally) was given for 5 hours. Rats were killed, serum prepared for determination of hepatocellular enzymes, and the liver assessed for iNOS and HO-1 by Western immunoblot. RESULTS LPS significantly increased serum aspartate aminotransferase levels, iNOS, and HO-1 immunoreactivity in the liver. Ketamine but not isoflurane attenuated LPS-induced liver injury, upregulated HO-1, and downregulated iNOS. CONCLUSION These data indicate that anesthetics differ in their effects on the liver in a rat model of sepsis with LPS. Ketamine has hepatoprotective effects against LPS-induced liver injury that appear to be mediated, at least in part, by differential modulation of the oxidative stress proteins iNOS and HO-1. Thus, ketamine may be the anesthetic agent of choice for septic patients requiring anesthesia.
Collapse
Affiliation(s)
- James W Suliburk
- Department of Surgery, The University of Texas Medical School at Houston, Houston, Texas, USA
| | | | | | | | | |
Collapse
|
18
|
Koksel O, Yildirim C, Cinel L, Tamer L, Ozdulger A, Bastürk M, Degirmenci U, Kanik A, Cinel I. Inhibition of poly(ADP-ribose) polymerase attenuates lung tissue damage after hind limb ischemia-reperfusion in rats. Pharmacol Res 2005; 51:453-62. [PMID: 15749460 DOI: 10.1016/j.phrs.2004.11.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/24/2004] [Indexed: 11/29/2022]
Abstract
The objective of this study was to investigate the effects of 3-aminobenzamide (3-AB) on tissue damage in lung after hind limb ischemia-reperfusion (I/R), by assessing blood biochemical assay and histopathological analysis. Thirty-five adult Wistar rats were divided into five groups. After application of anaesthesia both hind limbs were occluded with tourniquets. Following ischemia period for 60 min, the tourniquets were removed allowing reperfusion for 120 min. The IR group received 0.5 ml of saline while the IR+AB group received 3-AB (10 mgkg(-1) intraperitoneally). The IR+DMSO group was given 0.5 ml 10% DMSO 30 min before the removal of the tourniquets. The control group received 0.5 ml saline and the AB group received 0.5 ml 3-AB (10 mgkg(-1)) intraperitoneally. At the end of the reperfusion period, mid-line sternotomy was performed. Blood samples were taken with cardiac puncture. Bronchoalveolar lavage (BAL) of the left lung was performed with saline. Right lung was preserved for histopathological evaluation and biochemical examination. Lung tissue malondialdehyde (MDA) and 3-nitrotyrosine levels, myeloperoxidase and Na+/K+ ATP-ase activities, wet to dry weight ratios, and plasma and BAL fluid MDA levels were determined. Histopathological evaluation was performed, too. Hind limb IR caused significant increase in the lung tissue 3-NT to total tyrosine ratio (p = 0.014), wet to dry weight ratio (p = 0.000), MPO activity (p = 0.000), and MDA levels (p = 0.000). The animals treated with 3-AB showed a statistically significant decrease in these values (p < 0.05). Na+/K+ ATP-ase activity which was found to be decreased significantly with IR, returned to near normal levels with 3-AB treatment. Additionally, lung tissue injury in IR group characterized with moderate interstitial congestion and neutrophil infiltration, showed remarkable amelioration following 3-AB treatment. Our results strongly support the view that poly(ADP-ribose) polymerase (PARP) plays an important role in the inflammatory process in hind limb I/R-induced lung injury and as a PARP inhibitor, 3-AB seems to have a potential to treat this inflammatory injury.
Collapse
Affiliation(s)
- Oguz Koksel
- Department of Thoracic Surgery, Mersin University, School of Medicine, 33079 Mersin, Turkey.
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Md S, Moochhala SM, Siew Yang KL, Lu J, Anuar F, Mok P, Ng KC. The role of selective nitric oxide synthase inhibitor on nitric oxide and PGE2 levels in refractory hemorrhagic-shocked rats. J Surg Res 2005; 123:206-14. [PMID: 15680380 DOI: 10.1016/j.jss.2004.07.243] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2004] [Indexed: 01/22/2023]
Abstract
BACKGROUND The up-regulation of nitric oxide (NO) and cyclooxgenase-2 (COX-2) has been implicated in the pathophysiology of hemorrhagic shock. We examined the effects of aminoguanidine (AG), which is a known inducible nitric oxide synthase (iNOS) inhibitor, and NS-398, a known COX-2 inhibitor, in our rat model of refractory hemorrhagic shock (RHS). MATERIAL AND METHODS We measured tissue iNOS and COX-2 protein expression, brain and plasma nitrate/nitrite and prostaglandin E2 (PGE2) levels, plasma creatinine and glutamic oxalacetic transaminase (GOT) levels, quantified the histological damages in kidney, liver, lung, and brain, survival rate, and mean arterial blood pressure (MABP) in RHS rats. RESULTS Semiquantitative analysis of tissues showed iNOS protein was not detected in AG + RHS rats but was detected in normal saline and NS-398 RHS rats. Tissue COX-2 protein was not detected in AG and NS-398 RHS rats but was detected in normal saline + RHS rats. The levels of brain and plasma nitrate/nitrite and PGE2 and plasma creatinine and GOT were significantly lower in the AG + RHS rat group when compared with the normal saline RHS rat group. Histological examinations also showed a reduction in organ damage for AG + RHS rats when compared with treated RHS rats. AG + RHS rats showed significantly increased survival and MABP level when compared with treated RHS rats. CONCLUSION Our present findings suggest that NO produced by iNOS might result in organ damages. This in turn might lead to COX-2 up-regulation, and it increases the production of reactive oxygen species and toxic prostanoids. NO-mediated organ damage might be one way in which toxic products of COX-2 might further contribute to NO's deleterious effect in the later stages of RHS. It is therefore suggested that treatment of AG via inhibition of NO might contribute to improved physiological parameters and survival rates following RHS.
Collapse
Affiliation(s)
- Shirhan Md
- Department of Pharmacology, National University of Singapore
| | | | | | | | | | | | | |
Collapse
|
20
|
Madar Z, Kalet-Litman S, Stark AH. Inducible Nitric Oxide Synthase Activity and Expression in Liver and Hepatocytes of Diabetic Rats. Pharmacology 2005; 73:106-12. [PMID: 15528954 DOI: 10.1159/000081952] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2004] [Accepted: 07/30/2004] [Indexed: 01/02/2023]
Abstract
Inducible nitric oxide synthase (iNOS) is expressed by the liver in a number of physiological and pathophysiological conditions. The aim of this study was to investigate the relationship between the diabetic state, iNOS and oxidative stress in the rat liver and isolated hepatocytes. Hepatic iNOS expression and activity was measured in both healthy and streptozotocin-induced diabetic rats and determined in hepatocytes in the presence and absence of insulin. Cu/Zn superoxide dismutase (SOD) and phosphatidylinositol-3-kinase (PI3K) were also measured. In a separate experiment lasting 3 weeks, diabetic rats received either no treatment, two daily injections of insulin or aminoguanidine in the drinking water. Diabetes led to increased activity (45%) and expression (70%) of liver iNOS, an effect that was attenuated by insulin treatment both in vitro and in whole animals. Hepatocyte iNOS expression increased by 56%. Hepatic SOD expression was elevated in the diabetic state, but activity levels were similar to healthy controls. Insulin treatment in vivo led to increased enzyme activity but expression was not modified. Levels of PI3K protein were significantly lower in diabetic rats while insulin treatment markedly increased expression. Aminoguanidine did not inhibit hepatic iNOS in this study. Glycemic control via insulin administration was able to downregulate enhanced hepatic iNOS activity and expression in the liver observed in the diabetic state and improve SOD activity, responses that can potentially reduce the free radical damage associated with diabetes.
Collapse
Affiliation(s)
- Zecharia Madar
- The Hebrew University of Jerusalem, Faculty of Agricultural, Food and Environmental Quality Sciences, Rehovot, Israel.
| | | | | |
Collapse
|
21
|
Wang WW, Smith DLH, Zucker SD. Bilirubin inhibits iNOS expression and NO production in response to endotoxin in rats. Hepatology 2004; 40:424-33. [PMID: 15368447 DOI: 10.1002/hep.20334] [Citation(s) in RCA: 169] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The inducible isoform of heme oxygenase (HO), HO-1, has been shown to play an important role in attenuating tissue injury. Because HO-1 catalyzes the rate-limiting step in bilirubin synthesis, we examined the hypothesis that bilirubin is a key mediator of HO-1 cytoprotection, employing a rat model of endotoxemia. Bilirubin treatment resulted in improved survival and attenuated liver injury in response to lipopolysaccharide infusion. Serum levels of NO and tumor necrosis factor alpha, key mediators of endotoxemia, and hepatic inducible nitric oxide synthase (iNOS) expression were significantly lower in bilirubin-treated rodents versus control animals. Both intraperitoneal and local administration of bilirubin also was found to ameliorate hindpaw inflammation induced by the injection of lambda-carrageenan. Consistent with in vivo results, bilirubin significantly inhibited iNOS expression and suppressed NO production in lipopolysaccharide (LPS)-stimulated RAW 264.7 murine macrophages. In contrast, bilirubin treatment induced a threefold increase in LPS-mediated prostaglandin synthesis in the absence of significant changes in cyclooxygenase expression or activity, suggesting that bilirubin enhances substrate availability for eicosanoid synthesis. Bilirubin had no effect on LPS-mediated activation of nuclear factor kappaB or p38 mitogen-activated protein kinase, consistent with a nuclear factor kappaB-independent mechanism of action. Taken together, these data support a cytoprotective role for bilirubin that is mediated, at least in part, through the inhibition of iNOS expression and, potentially, through stimulation of local prostaglandin E2 production. In conclusion, our findings suggest a role for bilirubin in mollifying tissue injury in response to inflammatory stimuli and support the possibility that the phenomenon of "jaundice of sepsis" represents an adaptive physiological response to endotoxemia. Supplementary material for this article can be found on the HEPATOLOGY website (http://interscience.wiley.com/jpages/0270-9139/suppmat/index.html).
Collapse
Affiliation(s)
- Weizheng W Wang
- Division of Digestive Diseases, University of Cincinnati, Cincinnati, OH 45267-0595, USA
| | | | | |
Collapse
|
22
|
Farghali H, Canová N, Kucera T, Martínek J, Masek K. Nitric oxide synthase inhibitors modulate lipopolysaccharide-induced hepatocyte injury: dissociation between in vivo and in vitro effects. Int Immunopharmacol 2004; 3:1627-38. [PMID: 14555288 DOI: 10.1016/s1567-5769(03)00185-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Effects of endotoxemia-induced NO production on rat liver and hepatocytes in culture were investigated. Rats were treated intraperitoneally with saline, lipopolysaccharide (LPS, 10 mg/kg), L-nitroarginine methyl ester (L-NAME)+LPS, aminoguanidine (AG)+LPS, FK 506+LPS, S-nitroso-N-acetyl penicillamine (SNAP)+L-NAME+LPS and SNAP+FK 506+LPS. Mortality, hepatocyte viability and liver function test were estimated. Liver morphology was observed by light and electron microscopy. Hepatocyte cultures were treated with LPS, cytokine mixture (CM) with or without FK 506, L-NAME or AG. Hepatocyte function and inducible form of NOS (iNOS) expression were evaluated. Twenty-four hours after treatments with saline, LPS, L-NAME+LPS, AG+LPS, FK 506+LPS, SNAP+L-NAME+LPS and SNAP+FK 506+LPS, rat mortalities were 0%, 10%, 48%, 8%, 20%, 38% and 0%, and hepatocyte viabilities were 93+/-3%, 80+/-3%, 52+/-8%, 88+/-1%, 70+/-3%, 80+/-4% and 82+/-3%, respectively. AG+LPS or L-NAME+LPS administration was followed by excessive vacuolization of hepatocytes with lesions in the intermediary lobule zone characterized by features of secondary necrosis as a continuation of apoptotic processes. SNAP+L-NAME+LPS resulted in a well-preserved structure of central vein lobules with sparse signs of apoptosis. Treatment with LPS or CM increased iNOS expression in hepatocyte culture, which was inhibited by L-NAME, FK 506 or AG. AG reduced LPS-induced rise in alanine aminotransferase leakage. LPS-induced NO exerts cytoprotective effects in vivo, while LPS-induced NO in vitro appears to be toxic. Based on the data of this report, one cannot use in vitro results to predict in vivo responses to LPS-induced NO production. The pharmacological modulation of iNOS expression or NO production in vivo or in vitro, therefore, by the development of specific NO donors or inhibitors is promising for improvement of hepatocyte functions under the two experimental conditions, respectively.
Collapse
Affiliation(s)
- Hassan Farghali
- Institute of Pharmacology, 1st Faculty of Medicine, Charles University, Albertov 4, 128 00, 2, Prague, Czech Republic.
| | | | | | | | | |
Collapse
|
23
|
Giusti-Paiva A, Martinez MR, Felix JVC, da Rocha MJA, Carnio EC, Elias LLK, Antunes-Rodrigues J. Simvastatin Decreases Nitric Oxide Overproduction and Reverts the Impaired Vascular Responsiveness Induced by Endotoxic Shock in Rats. Shock 2004; 21:271-5. [PMID: 14770041 DOI: 10.1097/10.shk.0000115756.74059.ce] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Lipopolysaccharides (LPS) can be used to induce experimental endotoxic shock, which is characterized by a significant decrease in mean arterial pressure (MAP) and a decreased vasoconstrictor response that have been attributed to excessive nitric oxide production. Inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMG-CoA reductase), in addition to lowering serum cholesterol levels, exert many pleiotropic effects, including anti-inflammatory action. In the present study, we investigated the effect of simvastatin, an HMG-CoA reductase inhibitor, on the production of nitric oxide and the cardiovascular response to LPS. Male Wistar rats were pretreated with different doses of simvastatin (10, 20, 40, and 80 mg/kg, i.p.) or saline 20 min before i.v. injection of LPS (1.5 mg/kg) or saline (control). MAP was continuously recorded and nitrate plasma concentration was determined during the 6-h experimental session at 1-h intervals. The pressor response to phenylephrine (1 microg/kg) was evaluated before and 6 h after LPS administration. In the LPS-treated group, there was a time-dependent increase in nitrate plasma concentration (P<0.001), and this response was decreased in simvastatin pretreated rats (P<0.001). We also observed that LPS decreased the pressor response to phenylephrine (P<0.001), an effect that was reverted by simvastatin pretreatment (P<0.05). However, simvastatin did not modify the decrease of MAP induced by LPS. We concluded that simvastatin decreases nitrate plasma concentration in response to LPS and recovers vascular responsiveness during an experimental endotoxic shock. These data suggest the potential use of HMG-CoA reductase inhibitors as a coadjuvant in the treatment of septic shock.
Collapse
|
24
|
Ding H, Huang JA, Tong J, Yu X, Yu JP. Influence of Kupffer cells on hepatic signal transduction as demonstrated by second messengers and nuclear transcription factors. World J Gastroenterol 2003; 9:2519-22. [PMID: 14606088 PMCID: PMC4656532 DOI: 10.3748/wjg.v9.i11.2519] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To understand the influence of Kupffer cell (KC) on signal transduction pathways in the liver.
METHODS: To decrease selectively the number and function of KC, Kunming mice were ip injected with a single dose of gadolinium chloride (GdCl3, 20 mg•kg-1), the time-effect relationship assessment was performed after 1 d, 3 d and 6 d. sALT, sGST, liver glycogen content, phagocytic index, and expression of CD68 were assessed as the indexes of hepatotoxicity and functions of KC respectively, and morphology of KC was observed with transmission electron microscopy. Furthermore, cAMP, PGE2 level, nitric oxide (NO) content, and mRNA expression of NFkappaBp65, Erk1, STAT1 were examined.
RESULTS: GdCl3 could selectively cause apoptosis of KC and obvious reduction of KC’s activity, but no hepatotoxicity was observed. One day after KC blockade, NO, PGE2, cAMP contents in the liver were reduced 21.0%, 6.94-fold, 8.3%, respectively, and mRNA expression of NFkappaBp65 was decreased 3.0-fold. The change tendency of NO, PGE2, and cAMP contents and mRNA expression of NFkappaBp65 were concomitant with recovery of the functions of KC. The contents of NO, PEG2, cAMP were increased when the functions of KC was recovered. However, all of the changes could not return to the normal level except NO content after 6 d Gdcl3 treatment. No obvious changes were found in STAT1 and Erk1 mRNA expression in the present study.
CONCLUSION: Hepatic NO, PGE2, cAMP level and mRNA expression of NFkappaBp65 are closely related with the status of KC. It suggests that KC may play an important role in the cell to cell signal transduction in the liver.
Collapse
Affiliation(s)
- Hong Ding
- Department of Pharmacology, College of Pharmacy, Wuhan University, Wuhan 430072, Hubei Province, China.
| | | | | | | | | |
Collapse
|
25
|
N/A. N/A. Shijie Huaren Xiaohua Zazhi 2003; 11:1059-1063. [DOI: 10.11569/wcjd.v11.i7.1059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
26
|
Cinel I, Avlan D, Cinel L, Polat G, Atici S, Mavioglu I, Serinol H, Aksoyek S, Oral U. Ischemic preconditioning reduces intestinal epithelial apoptosis in rats. Shock 2003; 19:588-92. [PMID: 12785017 DOI: 10.1097/01.shk.0000055817.40894.84] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Recent experimental studies have described protective effect of ischemic preconditioning (IPC) on ischemia-reperfusion (I/R) injury of the intestine. We hypothesize that to reach a new point of view on the effect of IPC in intestinal barrier function, the relationship between I/R-induced mucosal injury and apoptosis must first be clarified. The present study was undertaken to investigate the role of IPC on intestinal apoptosis and probable contributions of bcl-2 expression to this process. We also investigated the effect of intestinal IPC on ileal malondyaldihyde levels. Forty-four male Wistar rats were randomized into four groups each consisting of 11 rats: sham-operated control, I/R group (30 min of superior mesenteric artery occlusion), IPC-I/R group (10 min of temporary artery occlusion prior before an ischemic insult of 30 min), and IPC alone group (10 min of preconditioning). Twenty-four hours later, ileum samples were obtained. Ileal malondyaldihyde levels were increased in the I/R group (31.9 +/- 18.8 vs. 106.8 +/- 39.8) but not in the IPC alone and IPC-I/R groups (38.1 +/- 13.6 and 44.7 +/- 12.7; P < 0.01). The number of apoptotic cells was significantly lower in IPC-I/R group than that of I/R group, and these findings were further supported by DNA laddering and M30 findings. Diminished bcl-2 expression observed in the ileal specimens of I/R group was prevented by IPC. Our results indicate that IPC may provide a protective effect on ileal epithelium and that this effect is probably the result of a significant increase in the expression of bcl-2 after the insult. The reversal of apoptosis by IPC might help preserving the vitality of intestinal structures that have a critical function, cessation of which often leads to multiorgan dysfunction syndrome.
Collapse
Affiliation(s)
- Ismail Cinel
- Department of Anesthesiology and Reanimation, Mersin University School of Medicine, Mersin, Turkey
| | | | | | | | | | | | | | | | | |
Collapse
|