1
|
Alchera E, Chandrashekar BR, Clemente N, Borroni E, Boldorini R, Carini R. Ischemia/Reperfusion Injury of Fatty Liver Is Protected by A2AR and Exacerbated by A1R Stimulation through Opposite Effects on ASK1 Activation. Cells 2021; 10:3171. [PMID: 34831394 PMCID: PMC8618984 DOI: 10.3390/cells10113171] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/11/2021] [Accepted: 11/13/2021] [Indexed: 12/11/2022] Open
Abstract
Hepatic ischemia/reperfusion injury (IRI) is aggravated by steatosis and is a main risk factor in fatty liver transplantation. Adenosine receptors (ARs) are emerging as therapeutic targets in liver diseases. By using cellular and in vivo systems of hepatic steatosis and IRI, here we evaluated the effects of pharmacological A2AR and A1R activation. The A2AR agonist CGS21680 protected the primary steatotic murine hepatocyte from IR damage and the activation of ASK1 and JNK. Such an effect was attributed to a phosphatidylinositol-3-kinase (PI3K)/Akt-dependent inhibition of ASK1. By contrast, the A1R agonist CCPA enhanced IR damage, intracellular steatosis and oxidative species (OS) production, thereby further increasing the lipid/OS-dependent ASK1-JNK stimulation. The CGS2680 and CCPA effects were nullified by a genetic ASK1 downregulation in steatotic hepatoma C1C7 cells. In steatotic mice livers, CGS21680 protected against hepatic IRI and ASK1/JNK activation whereas CCPA aggravated hepatic steatosis and IRI, and enhanced ASK1 and JNK stimulation. These results evidence a novel mechanism of CGS21680-mediated hepatoprotection, i.e., the PI3K/AKT-dependent inhibition of ASK1, and they show that CGS21680 and CCPA reduces and enhances the IRI of fatty liver, respectively, by preventing or increasing the activation of the cytotoxic ASK1/JNK axis. They also indicate the selective employment of A2AR agonists as an effective therapeutic strategy to prevent IRI in human fatty liver surgery.
Collapse
Affiliation(s)
| | | | | | | | | | - Rita Carini
- Department of Health Science, University of Piemonte Orientale, 28100 Novara, Italy; (E.A.); (B.R.C.); (N.C.); (E.B.); (R.B.)
| |
Collapse
|
2
|
Czigany Z, Craigie EC, Lurje G, Song S, Yonezawa K, Yamamoto Y, Minor T, Tolba RH. Adenosine A2a Receptor Stimulation Attenuates Ischemia-Reperfusion Injury and Improves Survival in A Porcine Model of DCD Liver Transplantation. Int J Mol Sci 2020; 21:E6747. [PMID: 32938013 PMCID: PMC7555737 DOI: 10.3390/ijms21186747] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 09/08/2020] [Accepted: 09/11/2020] [Indexed: 12/14/2022] Open
Abstract
Orthotopic liver transplantation (OLT) using allografts from donation after circulatory death (DCD) is potentially associated with compromised clinical outcomes due to ischemia-reperfusion injury (IRI)-induced organ damage and graft-related complications. The aim of this study was to provide in vivo data on the effects of adenosine A2a receptor stimulation in a clinically relevant large animal model of DCD liver transplantation. Cardiac arrest was induced in German Landrace pigs (n = 10; 20-25 kg). After 30 min of warm ischemia, the donor liver was retrieved following a cold flush with 3 L of histidine-tryptophan-ketoglutarate-HTK solution. Animals of the treatment group (n = 5/group) received a standard dose of the selective adenosine receptor agonist CGS 21680 added to the cold flush. All grafts were stored for 4.5 h at 4 °C in HTK-solution before OLT. Hepatocellular injury, apoptosis, protein kinase A-PKA activity, graft microcirculation, liver function, and animal survival were assessed. Compared to untreated livers, adenosine A2a receptor stimulation resulted in improved tissue microcirculation (103% ± 5% vs. 38% ± 4% compared to baseline; p < 0.05), accelerated functional recovery of the graft (indocyanine green-plasma disappearance rate (ICG-PDR) of 75% ± 18% vs. 40% ± 30% after 3 h), increased PKA activity ratio (56% ± 3% vs. 32% ± 3%; p < 0.001 after 1 h), and consequently reduced tissue necrosis and apoptosis. The potent protective effects were clinically manifested in significantly improved survival in the treatment group after 72 h (100% vs. 40%; p = 0.04). The ex vivo administration of adenosine A2a receptor agonist during the back-table flush mitigates IRI-mediated tissue damage and improves functional graft recovery and survival in a large animal model of DCD liver transplantation.
Collapse
Affiliation(s)
- Zoltan Czigany
- Department of Surgery and Transplantation, Faculty of Medicine, University Hospital RWTH Aachen, 52074 Aachen, Germany;
- Institute for Laboratory Animal Science and Experimental Surgery, Faculty of Medicine, RWTH-Aachen University, 52074 Aachen, Germany;
| | - Eve Christiana Craigie
- Institute for Laboratory Animal Science and Experimental Surgery, Faculty of Medicine, RWTH-Aachen University, 52074 Aachen, Germany;
| | - Georg Lurje
- Department of Surgery, Campus Charité Mitte | Campus Virchow-Klinikum–Charité-Universitätsmedizin, 13353 Berlin, Germany;
| | - Shaowei Song
- Department of Surgery, The First Affiliated Hospital of China Medical University, Shenyang 110122, China;
| | - Kei Yonezawa
- Department of Surgery, Shizuoka City Hospital, Shizuoka 420-8527, Japan;
| | - Yuzo Yamamoto
- Department of Gastroenterological Surgery, Akita University Graduate School of Medicine, Akita 010-0825, Japan;
| | - Thomas Minor
- Department of General, Visceral, and Transplantation Surgery, University Hospital Essen, 45147 Essen, Germany;
| | - René Hany Tolba
- Institute for Laboratory Animal Science and Experimental Surgery, Faculty of Medicine, RWTH-Aachen University, 52074 Aachen, Germany;
| |
Collapse
|
3
|
Li S, Yi Z, Deng M, Scott MJ, Yang C, Li W, Lei Z, Santerre NM, Loughran P, Billiar TR. TSLP protects against liver I/R injury via activation of the PI3K/Akt pathway. JCI Insight 2019; 4:129013. [PMID: 31723054 DOI: 10.1172/jci.insight.129013] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 10/10/2019] [Indexed: 01/09/2023] Open
Abstract
Thymic stromal lymphopoietin (TSLP) is a cytokine mainly released by epithelial cells that plays important roles in inflammation, autoimmune disease, and cancer. While TSLP is expressed in the liver at high levels, the role of TSLP in liver ischemia/reperfusion (I/R) injury remains unknown. Experiments were carried out to determine the role of TSLP in liver I/R injury. Wild-type (WT) and TSLP receptor-knockout (TSLPR-/-) mice were subjected to liver partial warm I/R injury. Liver injury was assessed by measuring serum alanine aminotransferase (ALT) level, necrotic areas by liver histology, hepatocyte death, and local hepatic inflammatory responses. Signal pathways were explored in vivo and in vitro to identify possible mechanisms for TSLP in I/R injury. TSLP and TSLPR protein expression increased during liver I/R in vivo and following hepatocyte hypoxia/reoxygenation in vitro. Deletion of TSLPR or neutralization of TSLP with anti-TSLP antibody exacerbated liver injury in terms of serum ALT levels as well as necrotic areas in liver histology. Administration of exogenous recombinant mouse TSLP to WT mice significantly reduced liver damage compared with controls, but failed to prevent I/R injury in TSLPR-/- mice. TSLP induced autophagy in hepatocytes during liver I/R injury. Mechanistically, Akt was activated in WT mice during liver I/R injury. The opposite results were observed in TSLPR-/- mice. In addition, TSLP could directly induce Akt activation in hepatocytes independent of nonparenchymal cells in vitro. Furthermore, the Akt agonist, insulin-like growth factor-1 (IGF-1), prevented I/R injury in TSLPR-/- mice and an Akt inhibitor, LY294002, blocked the protective effects of TSLP in WT mice subjected to I/R. Our data indicate that TSLP protects against liver I/R injury via activation of the PI3K/Akt pathway. Through this pathway, TSLP induces autophagy in hepatocytes. Thus, TSLP is a potent inhibitor of stress-induced hepatocyte necrosis.
Collapse
Affiliation(s)
- Shilai Li
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhongjie Yi
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Hepatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Meihong Deng
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Melanie J Scott
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Chenxuan Yang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,School of Medicine, Tsinghua University, Beijing, China
| | - Wenbo Li
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Plastic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhao Lei
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Hepatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Nicole M Santerre
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Patricia Loughran
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Center for Biologic Imaging, University of Pittsburgh, Pennsylvania, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
4
|
Gao Y, Zhou S, Wang F, Zhou Y, Sheng S, Qi D, Huang JH, Wu E, Lv Y, Huo X. Hepatoprotective effects of limb ischemic post-conditioning in hepatic ischemic rat model and liver cancer patients via PI3K/ERK pathways. Int J Biol Sci 2018; 14:2037-2050. [PMID: 30585267 PMCID: PMC6299361 DOI: 10.7150/ijbs.28435] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 10/05/2018] [Indexed: 12/11/2022] Open
Abstract
The most effective way of treating liver cancer is surgical resection, which usually requires blocking the hepatic portal circulation, and may result in hepatic ischemia-reperfusion injury (HIRI). It is of paramount importance to control HIRI for liver cancer surgical resection. In this study, a 70% ischemia-reperfusion (I/R) model of rat liver was established, and the protective effect and mechanism of limb ischemic post-conditioning (LIPOC) on HIRI was investigated. We show that LIPOC has a protective effect on hepatic ischemia-reperfusion injury in rats, which reduces the elimination of superoxide dismutase, thereby increasing oxygen free radical scavenging, decreasing lipid peroxidation, inhibiting neutrophil aggregation, as well as reducing TNFα, IL1β, and other inflammatory cytokines. In addition, LIPOC inhibited the apoptosis of hepatocytes induced by I/R injury, and decreased the Bax/Bcl-2 ratio. Furthermore, LIPOC promoted the phosphorylation of Akt and ERK1/2. The use of PI3K inhibitor LY294002 and ERK1/2 blocker PD98059 inhibited the phosphorylation of Akt and ERK1/2 caused by LIPOC and abolished the injury protection of liver I/R. Moreover, through 16 cases of hepatocellular carcinoma resections, we found that short-term LIPOC treatment significantly suppressed the elevated alanine aminotransferase, aspartic transaminase, and total bilirubin in the early post-operation of liver resection, and reduced reperfusion injury to the ischemic liver. In summary, our study demonstrates that LIPOC could be an effective method for HIRI in the clinical implementation of liver resection and uncovers the potential mechanism of LIPOC in the protective effects of HIRI.
Collapse
Affiliation(s)
- Yanfeng Gao
- Department of Anesthesiology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Shuang Zhou
- Department of Neurosurgery, Baylor Scott & White Health, Temple, Texas 76502, USA.,Neuroscience Institute, Baylor Scott & White Health, Temple, Texas 76502, USA
| | - Fengfei Wang
- Department of Neurosurgery, Baylor Scott & White Health, Temple, Texas 76502, USA.,Neuroscience Institute, Baylor Scott & White Health, Temple, Texas 76502, USA.,Department of Surgery, Texas A & M University Health Science Center, College of Medicine, Texas 76508, USA.,Department of Neurology, Baylor Scott & White Health, Temple, Texas 76502, USA
| | - Yue Zhou
- Department of Statistics, North Dakota State University, Fargo, North Dakota 58105, USA
| | - Sen Sheng
- Department of Neurology, University of Arkansas for Medical Science, Little Rock, Arkansas 72205, USA
| | - Dan Qi
- Department of Neurosurgery, Baylor Scott & White Health, Temple, Texas 76502, USA.,Neuroscience Institute, Baylor Scott & White Health, Temple, Texas 76502, USA
| | - Jason H Huang
- Department of Neurosurgery, Baylor Scott & White Health, Temple, Texas 76502, USA.,Neuroscience Institute, Baylor Scott & White Health, Temple, Texas 76502, USA.,Department of Surgery, Texas A & M University Health Science Center, College of Medicine, Texas 76508, USA
| | - Erxi Wu
- Department of Neurosurgery, Baylor Scott & White Health, Temple, Texas 76502, USA.,Neuroscience Institute, Baylor Scott & White Health, Temple, Texas 76502, USA.,Department of Surgery, Texas A & M University Health Science Center, College of Medicine, Texas 76508, USA.,Department of Pharmaceutical Sciences, Texas A & M University Health Science Center, College of Pharmacy, College Station, Texas 77843, USA.,LIVESTRONG Cancer Institutes, Dell Medical School, the University of Texas at Austin, Austin, Texas 78712, USA
| | - Yi Lv
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiongwei Huo
- Department of General Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
5
|
Novel Targets for Treating Ischemia-Reperfusion Injury in the Liver. Int J Mol Sci 2018; 19:ijms19051302. [PMID: 29701719 PMCID: PMC5983804 DOI: 10.3390/ijms19051302] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 04/20/2018] [Accepted: 04/24/2018] [Indexed: 12/22/2022] Open
Abstract
Liver ischemia-reperfusion injury (IRI) is a major complication of hemorrhagic shock, liver transplantation, and other liver surgeries. It is one of the leading causes for post-surgery hepatic dysfunction, always leading to morbidity and mortality. Several strategies, such as low-temperature reperfusion and ischemic preconditioning, are useful for ameliorating liver IRI in animal models. However, these methods are difficult to perform in clinical surgeries. It has been reported that the activation of peroxisome proliferator activated receptor gamma (PPARγ) protects the liver against IRI, but with unidentified direct target gene(s) and unclear mechanism(s). Recently, FAM3A, a direct target gene of PPARγ, had been shown to mediate PPARγ’s protective effects in liver IRI. Moreover, noncoding RNAs, including LncRNAs and miRNAs, had also been reported to play important roles in the process of hepatic IRI. This review briefly discussed the roles and mechanisms of several classes of important molecules, including PPARγ, FAM3A, miRNAs, and LncRNAs, in liver IRI. In particular, oral administration of PPARγ agonists before liver surgery or liver transplantation to activate hepatic FAM3A pathways holds great promise for attenuating human liver IRI.
Collapse
|
6
|
He B, Hoang TK, Tran DQ, Rhoads JM, Liu Y. Adenosine A 2A Receptor Deletion Blocks the Beneficial Effects of Lactobacillus reuteri in Regulatory T-Deficient Scurfy Mice. Front Immunol 2017; 8:1680. [PMID: 29270168 PMCID: PMC5723640 DOI: 10.3389/fimmu.2017.01680] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 11/15/2017] [Indexed: 01/06/2023] Open
Abstract
The lack of a functional Foxp3 transcription factor and regulatory T (Treg) cells causes lethal, CD4+ T cell-driven autoimmune diseases in scurfy (SF) mice and humans. Recent studies have shown that adenosine A2A receptor activation limits inflammation and tissue damage, thereby playing an anti-inflammatory role. However, the role of the adenosine A2A receptor in the development of disease in SF mice remains unclear. Using a genetic approach, we found that adenosine A2A receptor deletion in SF mice (SF[Formula: see text]) does not affect early life events, the development of a lymphoproliferative disorder, or hyper-production of pro-inflammatory cytokines seen in the Treg-deficiency state. As shown previously, Lactobacillus reuteri DSM 17938 treatment prolonged survival and reduced multiorgan inflammation in SF mice. In marked contrast, A2A receptor deletion completely blocked these beneficial effects of L. reuteri in SF mice. Altogether, these results suggest that although absence of the adenosine A2A receptor does not affect the development of disease in SF mice, it plays a critical role in the immunomodulation by L. reuteri in Treg-deficiency disease. The adenosine A2A receptor and its activation may have a role in treating other Treg dysfunction-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Baokun He
- Division of Gastroenterology, Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Thomas K. Hoang
- Division of Gastroenterology, Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Dat Q. Tran
- Division of Gastroenterology, Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jon Marc Rhoads
- Division of Gastroenterology, Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Yuying Liu
- Division of Gastroenterology, Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
7
|
Jiménez-Castro MB, Negrete-Sánchez E, Casillas-Ramírez A, Gulfo J, Álvarez-Mercado AI, Cornide-Petronio ME, Gracia-Sancho J, Rodés J, Peralta C. The effect of cortisol in rat steatotic and non-steatotic liver transplantation from brain-dead donors. Clin Sci (Lond) 2017; 131:733-746. [PMID: 28246131 DOI: 10.1042/cs20160676] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 02/13/2017] [Accepted: 02/27/2017] [Indexed: 12/22/2022]
Abstract
In the present study, we examined the effects of cortisol on steatotic and non-steatotic liver grafts from brain-dead donors and characterized the underlying mechanisms involved. Non-steatotic liver grafts showed reduced cortisol and increased cortisone levels in association with up-regulation of enzymes that inactivate cortisol. Conversely, steatotic liver grafts exhibited increased cortisol and reduced cortisone levels. The enzymes involved in cortisol generation were overexpressed, and those involved in cortisol inactivation or clearance were down-regulated in steatotic liver grafts. Exogenous administration of cortisol negatively affected hepatic damage and survival rate in non-steatotic liver transplantation (LT); however, cortisol treatment up-regulated the phosphoinositide 3-kinase (PI3K)-protein kinase C (PKC) pathway, resulting in protection against the deleterious effects of brain-dead donors on damage and inflammatory response in steatotic LT as well as in increased survival of recipients. The present study highlights the differences in the role of cortisol and hepatic mechanisms that regulate cortisol levels based on the type of liver. Our findings suggest that cortisol treatment is a feasible and highly protective strategy to reduce the adverse effects of brain-dead donor livers in order to ultimately improve liver graft quality in the presence of steatosis, whereas cortisol treatment would not be recommended for non-steatotic liver grafts.
Collapse
Affiliation(s)
| | | | - Araní Casillas-Ramírez
- Hospital Regional de Alta Especialidad de Ciudad Victoria, México
- Facultad de Medicina e Ingeniería en Sistemas Computacionales de Matamoros, Universidad Autónoma de Tamaulipas, México
| | - Jose Gulfo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona, Spain
| | | | | | - Jordi Gracia-Sancho
- Barcelona Hepatic Hemodynamic Laboratory, IDIBAPS, CIBEREHD, Barcelona, Spain
| | - Juan Rodés
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)
- Liver Transplant Unit, Hospital Clínic, Universitat de Barcelona, IDIBAPS, Spain
| | - Carmen Peralta
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona, Spain
| |
Collapse
|
8
|
Inhalation of high concentrations of hydrogen ameliorates liver ischemia/reperfusion injury through A 2A receptor mediated PI3K-Akt pathway. Biochem Pharmacol 2017; 130:83-92. [DOI: 10.1016/j.bcp.2017.02.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 02/01/2017] [Indexed: 12/12/2022]
|
9
|
Alchera E, Rolla S, Imarisio C, Bardina V, Valente G, Novelli F, Carini R. Adenosine A2a receptor stimulation blocks development of nonalcoholic steatohepatitis in mice by multilevel inhibition of signals that cause immunolipotoxicity. Transl Res 2017; 182:75-87. [PMID: 28011152 DOI: 10.1016/j.trsl.2016.11.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 11/09/2016] [Accepted: 11/26/2016] [Indexed: 12/22/2022]
Abstract
Lipotoxicity and immunoinflammation are associated with the evolution of steatosis toward nonalcoholic steatohepatitis (NASH). This study reports the ability of adenosine A2a receptor (A2aR) activation to inhibit NASH development by modulating the responses of CD4+ T-helper (Th) cells to avoid an immuno-mediated potentiation of lipotoxicity. The effect of the A2aR agonist CGS21680 on immunoinflammatory signals, CD4+Th cell infiltration and immunolipotoxicity was analyzed in steatotic C57BL/6 mice fed with a methionine-choline-deficient (MCD) diet and in mouse hepatocytes exposed to palmitic acid (PA). CGS21680 inhibited NASH development in steatotic mice and decreased cytokines and chemokines involved in Th cell recruitment or polarization (namely CXCL10, CCL2, tumor necrosis factor alfa [TNFα], tumor growth factor [TGFβ], and IL-12). CGS21680 also reduced the expansion of Th17, Th22, and Th1 cells and increased the immunosuppressive activity of T regulatory cells. In PA-treated mice hepatocytes, CGS21680 inhibited the production of CXCL10, TNFα, TGFβ, IL-12, and CCL2; CGS21680 also prevented JNK-dependent lipotoxicity and its intensification by IL-17 or IL-17 plus IL-22 through Akt/PI3-kinase stimulation and inhibition of the negative regulator of PI3-kinase, (phosphatase and tensin homologue deleted from chromosome 10 (PTEN), which is upregulated by IL-17. In MCD livers, CGS21680 reduced JNK activation and PTEN expression and increased Akt phosphorylation. In conclusion, A2aR stimulation inhibited NASH development by reducing Th17 cell expansion and inhibiting the exacerbation of the IL-17-induced JNK-dependent lipotoxicity. These data promote the implementation of further studies to evaluate the potential clinical application of A2aR agonists that, by being able to function as both cytoprotective and immunomodulatory agents, could efficiently antagonize the multi-faced pathogenesis of NASH.
Collapse
Affiliation(s)
- Elisa Alchera
- Department of Health Science, University of Piemonte Orientale, Novara, Italy
| | - Simona Rolla
- Center for Experimental Research and Medical Studies (CERMS), Azienda Ospedaliera Città della Salute e della Scienza di Torino, Torino, Italy; Department of Clinical and Biological Sciences, University of Turin, Orbassano, Italy
| | - Chiara Imarisio
- Department of Health Science, University of Piemonte Orientale, Novara, Italy
| | - Valentina Bardina
- Center for Experimental Research and Medical Studies (CERMS), Azienda Ospedaliera Città della Salute e della Scienza di Torino, Torino, Italy; Department of Clinical and Biological Sciences, University of Turin, Orbassano, Italy
| | - Guido Valente
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Francesco Novelli
- Center for Experimental Research and Medical Studies (CERMS), Azienda Ospedaliera Città della Salute e della Scienza di Torino, Torino, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy; Immunogenetics and Transplantation Biology Unit, Azienda Ospedaliera Città della Salute e della Scienza di Torino, Turin, Italy
| | - Rita Carini
- Department of Health Science, University of Piemonte Orientale, Novara, Italy.
| |
Collapse
|
10
|
Covington SM, Bauler LD, Toledo-Pereyra LH. Akt: A Therapeutic Target in Hepatic Ischemia-Reperfusion Injury. J INVEST SURG 2016; 30:47-55. [PMID: 27463073 DOI: 10.1080/08941939.2016.1206999] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Liver transplantation is the second most common transplant procedure in the United States. A leading cause of post-transplantation organ dysfunction is I/R injury. During I/R injury, the serine/threonine kinase Akt is activated, stimulating downstream mediators to promote cellular survival. Due to the cellular effects of Akt, therapeutic manipulation of the Akt pathway can help reduce cellular damage during hepatic I/R that occurs during liver transplantation. OBJECTIVE A full description of therapeutic options available that target Akt to reduce hepatic I/R injury has not been addressed within the literature. The purpose of this review is to illuminate advances in the manipulation of Akt that can be used to therapeutically target I/R injury in the liver. METHODS An in depth literature review was performed using the Scopus and PubMed databases. A total of 75 published articles were utilized for this manuscript. Terminology searched includes a combination of "hepatic ischemia/reperfusion injury", "Akt/PKB", "preconditioning" and "postconditioning." RESULTS Four principal methods that reduce I/R injury include hepatic pre- and postconditioning, pharmacological intervention and future miRNA/gene therapy. Discussed therapies used serum alanine aminotransferase levels, liver histology and phosphorylation of downstream mediators to confirm the Akt protective effect. CONCLUSION The activation of Akt from the reviewed therapies has resulted in predictable reduction in hepatocyte damage using the previously mentioned measurements. In a clinical setting, these therapies could potentially be used in combination to achieve better outcomes in hepatic transplant patients. Evidence supporting reduced I/R injury through Akt activation warrants further studies in human clinical trials.
Collapse
Affiliation(s)
- Stephen M Covington
- a Michigan State University College of Osteopathic Medicine , East Lansing, Michigan , USA
| | - Laura D Bauler
- b Division of Epidemiology and Biostatistics , Western Michigan University Homer Stryker M.D. School of Medicine , Kalamazoo , Michigan , USA
| | - Luis H Toledo-Pereyra
- b Division of Epidemiology and Biostatistics , Western Michigan University Homer Stryker M.D. School of Medicine , Kalamazoo , Michigan , USA
| |
Collapse
|
11
|
Iskandarov E, Kadaba Srinivasan P, Xin W, Bleilevens C, Afify M, Hamza A, Wei L, Hata K, Agayev B, Tolba R. Protective Effects of Adenosine Receptor Agonist in a Cirrhotic Liver Resection Model. HEPATITIS MONTHLY 2016; 16:e36821. [PMID: 27799962 PMCID: PMC5075226 DOI: 10.5812/hepatmon.36821] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 03/07/2016] [Accepted: 06/22/2016] [Indexed: 12/11/2022]
Abstract
OBJECTIVES To investigate the role of CGS21680, a selective adenosine A2A receptor agonist, on a bile-duct-ligated cirrhotic liver resection model in rats. METHODS Male Wistar rats were allotted into 3 groups (n = 7 per time-point): the control group, the bile duct ligation + CGS21680 group (BDL + CGS), and the bile duct ligation group (BDL). Biliary cirrhosis had been previously induced by ligature of the common bile duct in the BDL + CGS and BDL groups. After 2 weeks, the animals underwent partial hepatectomy (50%). The BDL + CGS group received a single dose of CGS21680 15 minutes prior to hepatectomy. Blood samples were collected and analyzed. RESULTS Aspartate transaminase levels were found to be lower in the control vs BDL groups (1, 3, and 24 h) (P < 0.01) and the BDL + CGS (1 and 3 hours) (P < 0.01) and BDL + CGS vs BDL (24 hours) (P < 0.05) groups. Hepatic flow was measured and BDL showed significantly lower values at the 3, 24, and 168 h time-points compared to the control (P < 0.01) and BDL + CGS groups (P < 0.05 at 3 and 168 hours; P < 0.01 at 24 h). O2C velocity was reduced in the BDL compared to the control group (P < 0.001 at 3 hours; P < 0.01 at 24 and 168 hours) and the BDL + CGS group (P < 0.01 at 24 hours). Interleukin-6 levels were abrogated in the BDL + CGS (P < 0.05) and control (P < 0.01) groups versus BDL. Histone-bound low-molecular-weight DNA fragments in the BDL + CGS (P < 0.01) and control (P < 0.05) groups were low compared to the BDL group. CONCLUSIONS Administration of CGS21680, an adenosine receptor agonist, after the resection of bile-duct-ligated cirrhotic livers led to improved liver function, regeneration, and microcirculation.
Collapse
Affiliation(s)
- Emil Iskandarov
- Department of Hepato-biliary and Pancreas Surgery, Scientific Center of Surgery named after academician M.A.Topchubashov, Baku, Azerbaijan
| | - Pramod Kadaba Srinivasan
- Institute for Laboratory Animal Science and Experimental Surgery, University Hospital, RWTH Aachen University, Aachen, Germany
- Corresponding Author: Pramod Kadaba Srinivasan, Institute for Laboratory Animal Science and Experimental Surgery, University Hospital RWTH Aachen, Aachen, Germany. Tel: +49-2418089855, Fax: +49-2418082462, E-mail:
| | - Wang Xin
- Tongji Hospital, Tongji Medical College, Department of Surgery, Division of Transplantation, Wuhan, China
| | - Christian Bleilevens
- Department of Anaesthesiology, University Hospital, RWTH Aachen University, Aachen, Germany
| | - Mamdouh Afify
- Institute for Laboratory Animal Science and Experimental Surgery, University Hospital, RWTH Aachen University, Aachen, Germany
| | - Astrit Hamza
- Department of Abdominal Surgery, University Clinical Center of Kosovo, Prishtina, Kosovo
| | - Lai Wei
- Tongji Hospital, Tongji Medical College, Department of Surgery, Division of Transplantation, Wuhan, China
| | - Koichiro Hata
- Department of Hepatobiliary, Pancreas and Transplant Surgery, Kyoto University, Yoshidahonmachi, Sakyo Ward, Kyoto, Japan
| | - Boyukkishi Agayev
- Department of Hepato-biliary and Pancreas Surgery, Scientific Center of Surgery named after academician M.A.Topchubashov, Baku, Azerbaijan
| | - Rene Tolba
- Institute for Laboratory Animal Science and Experimental Surgery, University Hospital, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
12
|
Beyond Preconditioning: Postconditioning as an Alternative Technique in the Prevention of Liver Ischemia-Reperfusion Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:8235921. [PMID: 27340509 PMCID: PMC4909928 DOI: 10.1155/2016/8235921] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 04/11/2016] [Accepted: 05/05/2016] [Indexed: 01/04/2023]
Abstract
Liver ischemia/reperfusion injury may significantly compromise hepatic postoperative function. Various hepatoprotective methods have been improvised, aiming at attenuating IR injury. With ischemic preconditioning (IPC), the liver is conditioned with a brief ischemic period followed by reperfusion, prior to sustained ischemia. Ischemic postconditioning (IPostC), consisting of intermittent sequential interruptions of blood flow in the early phase of reperfusion, seems to be a more feasible alternative than IPC, since the onset of reperfusion is more predictable. Regarding the potential mechanisms involved, it has been postulated that the slow intermittent oxygenation through controlled reperfusion decreases the burst production of oxygen free radicals, increases antioxidant activity, suppresses neutrophil accumulation, and modulates the apoptotic cascade. Additionally, favorable effects on mitochondrial ultrastructure and function, and upregulation of the cytoprotective properties of nitric oxide, leading to preservation of sinusoidal structure and maintenance of blood flow through the hepatic circulation could also underlie the protection afforded by postconditioning. Clinical studies are required to show whether biochemical and histological improvements afforded by the reperfusion/reocclusion cycles of postconditioning during early reperfusion can be translated to a substantial clinical benefit in liver resection and transplantation settings or to highlight more aspects of its molecular mechanisms.
Collapse
|
13
|
Shaikh G, Cronstein B. Signaling pathways involving adenosine A2A and A2B receptors in wound healing and fibrosis. Purinergic Signal 2016; 12:191-7. [PMID: 26847815 DOI: 10.1007/s11302-016-9498-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 01/27/2016] [Indexed: 02/07/2023] Open
Abstract
Collagen and matrix deposition by fibroblasts is an essential part of wound healing but also contributes to pathologic remodeling of organs leading to substantial morbidity and mortality. Adenosine, a small molecule generated extracellularly from adenine nucleotides as a result of direct stimulation, hypoxia, or injury, acts via a family of classical seven-pass G protein-coupled protein receptors, A2A and A2B, leading to generation of cAMP and activation of downstream targets such as PKA and Epac. These effectors, in turn, lead to fibroblast activation and collagen synthesis. The regulatory actions of these receptors likely involve multiple interconnected pathways, and one of the more interesting aspects of this regulation is opposing effects at different levels of cAMP generated. Additionally, adenosine signaling contributes to fibrosis in organ-specific ways and may have opposite effects in different organs. The development of drugs that selectively target these receptors and their signaling pathways will disrupt the pathogenesis of fibrosis and slow or arrest the progression of the important diseases they underlie.
Collapse
Affiliation(s)
- Gibran Shaikh
- Department of Medicine, New York University School of Medicine, 227 East 30th Street, New York, NY, 10016, USA
| | - Bruce Cronstein
- Department of Medicine, New York University School of Medicine, 227 East 30th Street, New York, NY, 10016, USA.
| |
Collapse
|
14
|
Pharmacological Preconditioning by Adenosine A2a Receptor Stimulation: Features of the Protected Liver Cell Phenotype. BIOMED RESEARCH INTERNATIONAL 2015; 2015:286746. [PMID: 26539478 PMCID: PMC4619783 DOI: 10.1155/2015/286746] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 04/15/2015] [Indexed: 02/06/2023]
Abstract
Ischemic preconditioning (IP) of the liver by a brief interruption of the blood flow protects the damage induced by a subsequent ischemia/reperfusion (I/R) preventing parenchymal and nonparenchymal liver cell damage. The discovery of IP has shown the existence of intrinsic systems of cytoprotection whose activation can stave off the progression of irreversible tissue damage. Deciphering the molecular mediators that underlie the cytoprotective effects of preconditioning can pave the way to important therapeutic possibilities. Pharmacological activation of critical mediators of IP would be expected to emulate or even to intensify its salubrious effects. In vitro and in vivo studies have demonstrated the role of the adenosine A2a receptor (A2aR) as a trigger of liver IP. This review will provide insight into the phenotypic changes that underline the resistance to death of liver cells preconditioned by pharmacological activation of A2aR and their implications to develop innovative strategies against liver IR damage.
Collapse
|
15
|
Zhang R, Zhang L, Manaenko A, Ye Z, Liu W, Sun X. Helium preconditioning protects mouse liver against ischemia and reperfusion injury through the PI3K/Akt pathway. J Hepatol 2014; 61:1048-55. [PMID: 24972044 DOI: 10.1016/j.jhep.2014.06.020] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 05/24/2014] [Accepted: 06/14/2014] [Indexed: 01/12/2023]
Abstract
BACKGROUND & AIMS Hepatic ischemia and reperfusion (I/R) injury is a major complication of liver transplantation, hepatic resection and trauma. Helium preconditioning (HPC) exerts protection against ischemic stress. We investigated potential beneficial effects of HPC on I/R-induced liver injury and investigated mechanisms underlying HPC-induced protection. METHODS We employed a model of segmental warm hepatic I/R on BALB/c mice. Serum ALT was measured and livers were analysed by histology, RT-PCR and western blot. HPC was induced by inhalation of a 70% helium/30% oxygen mixture for three 5-min periods, interspersed with three 5-min washout periods by room air. We tested which component of HPC (the helium/air mixture inhalation, the air room gap, or the interaction between these two factors) is protective. RESULTS We found that HPC caused a significant increase in Akt phosphorylation in hepatocytes. The HPC-induced Akt phosphorylation resulted in decreased hepatocellular injury and improved survival rate of the treated animals. PI3K inhibitors abolished HPC induced effects. HPC-induced Akt phosphorylation affected expression of its downstream molecules. The effects of HPC on the PI3K/Akt pathway were attenuated by adenosine A2A receptor blockade, but could be re-established by PTEN inhibition. We demonstrated that the interaction of helium/air breathing and air gaps is responsible for the observed effects of HPC. CONCLUSIONS HPC may be a promising strategy leading to a decrease in I/R induced liver injury in clinical settings. Additionally, the PI3K/Akt pathway plays an essential role in the protective effects of HPC in hepatic I/R injury.
Collapse
Affiliation(s)
- Rongjia Zhang
- Department of Diving Medicine, Second Military Medical University, Shanghai, China
| | - Ling Zhang
- Department of Medical Genetics, Second Military Medical University, Shanghai, China
| | - Anatol Manaenko
- Department of Physiology and Pharmacology, Loma Linda University Medical Center, Loma Linda, CA, USA
| | - Zhouheng Ye
- Department of Diving Medicine, Second Military Medical University, Shanghai, China
| | - Wenwu Liu
- Department of Diving Medicine, Second Military Medical University, Shanghai, China.
| | - Xuejun Sun
- Department of Diving Medicine, Second Military Medical University, Shanghai, China.
| |
Collapse
|
16
|
Yue S, Rao J, Zhu J, Busuttil RW, Kupiec-Weglinski JW, Lu L, Wang X, Zhai Y. Myeloid PTEN deficiency protects livers from ischemia reperfusion injury by facilitating M2 macrophage differentiation. THE JOURNAL OF IMMUNOLOGY 2014; 192:5343-5353. [PMID: 24771857 DOI: 10.4049/jimmunol.1400280] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Although the role of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) in regulating cell proliferation is well established, its function in immune responses remains to be fully appreciated. In the current study, we analyzed myeloid-specific PTEN function in regulating tissue inflammatory immune response in a murine liver partial warm ischemia model. Myeloid-specific PTEN knockout (KO) resulted in liver protection from ischemia reperfusion injury (IRI) by deviating the local innate immune response against ischemia reperfusion toward the regulatory type: expression of proinflammatory genes was selectively decreased and anti-inflammatory IL-10 was simultaneously increased in ischemia reperfusion livers of PTEN KO mice compared with those of wild-type (WT) mice. PI3K inhibitor and IL-10-neutralizing Abs, but not exogenous LPS, recreated liver IRI in these KO mice. At the cellular level, Kupffer cells and peritoneal macrophages isolated from KO mice expressed higher levels of M2 markers and produced lower TNF-α and higher IL-10 in response to TLR ligands than did their WT counterparts. They had enhanced Stat3- and Stat6-signaling pathway activation, but diminished Stat1-signaling pathway activation, in response to TLR4 stimulation. Inactivation of Kupffer cells by gadolinium chloride enhanced proinflammatory immune activation and increased IRI in livers of myeloid PTEN KO mice. Thus, myeloid PTEN deficiency protects livers from IRI by facilitating M2 macrophage differentiation.
Collapse
Affiliation(s)
- Shi Yue
- 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
| | - Jianhua Rao
- 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.,Department of Liver Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiansu Province, China
| | - Jianjun Zhu
- 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.,Department of Liver Surgery, Renji Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Ronald W Busuttil
- 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
| | - Jerzy W Kupiec-Weglinski
- 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
| | - Ling Lu
- Department of Liver Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiansu Province, China
| | - Xuehao Wang
- Department of Liver Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiansu Province, China
| | - Yuan Zhai
- 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
| |
Collapse
|
17
|
Grossini E, Pollesello P, Bellofatto K, Sigaudo L, Farruggio S, Origlia V, Mombello C, Mary DASG, Valente G, Vacca G. Protective effects elicited by levosimendan against liver ischemia/reperfusion injury in anesthetized rats. Liver Transpl 2014; 20:361-75. [PMID: 24273004 DOI: 10.1002/lt.23799] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 11/09/2013] [Indexed: 02/06/2023]
Abstract
As in other organs, oxidative stress-induced injury and cell death may result from free oxygen radical-dependent mechanisms and alterations in signal transduction pathways leading to apoptosis. Among the new suggested therapies for injuries caused by oxidative stress, the use of levosimendan has been reported to be quite promising. In the present study, we aimed to examine the protective effects of levosimendan against liver oxidative stress in anesthetized rats and to analyze the involvement of mitochondrial adenosine triphosphate-dependent potassium (mitoK(ATP)) channels and nitric oxide (NO). In 50 anesthetized rats, liver ischemia/reperfusion (I/R) was performed via nontraumatic portal occlusion. In some animals, levosimendan was infused into the portal vein at the onset of reperfusion, whereas other rats received the vehicle only. Moreover, in some rats, levosimendan was given after the intraportal administration of L-Nω-nitro-arginine methyl ester (L-NAME) or 5-hydroxydecanoate (5HD). The portal vein blood flow was measured, and blood samples were taken for the determination of transaminases, thiobarbituric acid reactive substances (TBARS), and reduced glutathione (GSH); liver biopsy samples were used for B cell lymphoma 2-associated X protein, caspase-9, Akt, and endothelial nitric oxide synthase (eNOS) activation through western blotting. Also, caspase-3 activity was measured. In rats, I/R caused an increase in apoptotic markers, transaminases, and TBARS and a decrease in GSH and Akt activation. Levosimendan administration was able to counteract oxidative damage and apoptosis in a dose-dependent way and to increase GSH, Akt, and eNOS activation. All effects of levosimendan were abolished by pretreatment with L-NAME and 5HD. In conclusion, the results of the present study show that levosimendan can exert protection against ischemic liver damage through mechanisms related to NO production and mitoKATP channel function. These data provide interesting perspectives into the use of levosimendan in hepatic surgery and transplantation.
Collapse
Affiliation(s)
- Elena Grossini
- Physiology Laboratory, Department of Translational Medicine, A. Avogadro University of East Piedmont, Novara, Italy; Experimental Surgery, Azienda Ospedaliera Universitaria Maggiore della Carità, Novara, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Baldanzi G. Inhibition of diacylglycerol kinases as a physiological way to promote diacylglycerol signaling. Adv Biol Regul 2014; 55:39-49. [PMID: 24582387 DOI: 10.1016/j.jbior.2014.02.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 01/02/2014] [Accepted: 02/02/2014] [Indexed: 01/12/2023]
Abstract
Diacylglycerol is a key regulator of cell physiology, controlling the membrane recruitment and activation of signaling molecules. Accordingly, diacylglycerol generation and metabolism are strictly controlled, allowing for localized regulation of its concentration. While the increased production of diacylglycerol upon receptor triggering is well recognized, the modulation of diacylglycerol metabolism by diacylglycerol kinases (DGKs) is less characterized. Some agonists induce DGK activation and recruitment to the plasma membrane, promoting diacylglycerol metabolism to phosphatidic acid. Conversely, several reports indicate that signaling pathways that selectively inhibits DGK isoforms can enhance cellular diacylglycerol levels and signal transduction. For example, the impairment of DGKθ activity by RhoA binding to the catalytic domain represents a conserved mechanism controlling diacylglycerol signaling from Caenorhabditis elegans motoneurons to mammalian hepatocytes. Similarly, DGKα activity is inhibited in lymphocytes by TCR signaling, thus contributing to a rise in diacylglycerol concentration for downstream signaling. Finally, DGKμ activity is inhibited by ischemia-reperfusion-generated reactive oxygen species in airway endothelial cells, promoting diacylglycerol-mediated ion channel opening and edema. In those systems, DGKs provide a gatekeeper function by blunting diacylglycerol levels or possibly establishing permissive domains for diacylglycerol signaling. In this review, I discuss the possible general relevance of DGK inhibition to enhanced diacylglycerol signaling.
Collapse
Affiliation(s)
- Gianluca Baldanzi
- University "A. Avogadro" del Piemonte Orientale, Department of Translational Medicine, via Solaroli 17, 28100 Novara, Italy.
| |
Collapse
|
19
|
Chen Z, Jia S, Li D, Cai J, Tu J, Geng B, Guan Y, Cui Q, Yang J. Silencing of long noncoding RNA AK139328 attenuates ischemia/reperfusion injury in mouse livers. PLoS One 2013; 8:e80817. [PMID: 24312245 PMCID: PMC3842297 DOI: 10.1371/journal.pone.0080817] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 10/03/2013] [Indexed: 11/18/2022] Open
Abstract
Recently, increasing evidences had suggested that long noncoding RNAs (LncRNAs) are involved in a wide range of physiological and pathophysiological processes. Here we determined the LncRNA expression profile using microarray technology in mouse livers after ischemia/reperfusion treatment. Seventy one LncRNAs were upregulated, and 27 LncRNAs were downregulated in ischemia/reperfusion-treated mouse livers. Eleven of the most significantly deregulated LncRNAs were further validated by quantitative PCR assays. Among the upregulated LncRNAs confirmed by quantitative PCR assays, AK139328 exhibited the highest expression level in normal mouse livers. siRNA-mediated knockdown of hepatic AK139328 decreased plasma aminotransferase activities, and reduced necrosis area in the livers with a decrease in caspase-3 activation after ischemia/reperfusion treatment. In ischemia/reperfusion liver, knockdown of AK139328 increased survival signaling proteins including phosphorylated Akt (pAkt), glycogen synthase kinase 3 (pGSK3) and endothelial nitric oxide synthase (peNOS). Furthermore, knockdown of AK139328 also reduced macrophage infitration and inhibited NF-κB activity and inflammatory cytokines expression. In conclusion, these findings revealed that deregulated LncRNAs are involved in liver ischemia/reperfusion injury. Silencing of AK139328 ameliorated ischemia/reperfusion injury in the liver with the activation of Akt signaling pathway and inhibition of NF-κB activity. LncRNA AK139328 might be a novel target for diagnosis and treatment of liver surgery or transplantation.
Collapse
Affiliation(s)
- Zhenzhen Chen
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Beijing, China
- MOE Key Laboratory of Molecular Cardiovascular Science, Peking University, Beijing, China
| | - Shi Jia
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Beijing, China
- MOE Key Laboratory of Molecular Cardiovascular Science, Peking University, Beijing, China
| | - Danhua Li
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Beijing, China
- MOE Key Laboratory of Molecular Cardiovascular Science, Peking University, Beijing, China
| | - Junyan Cai
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Beijing, China
- MOE Key Laboratory of Molecular Cardiovascular Science, Peking University, Beijing, China
| | - Jian Tu
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Beijing, China
- Department of Biomedical Informatics, Peking University School of Basic Medical Sciences, Beijing, China
| | - Bin Geng
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Beijing, China
- MOE Key Laboratory of Molecular Cardiovascular Science, Peking University, Beijing, China
| | - Youfei Guan
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Beijing, China
- MOE Key Laboratory of Molecular Cardiovascular Science, Peking University, Beijing, China
| | - Qinghua Cui
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Beijing, China
- Department of Biomedical Informatics, Peking University School of Basic Medical Sciences, Beijing, China
- Institute of Systems Biomedicine, Peking University, Beijing, China
- MOE Key Laboratory of Molecular Cardiovascular Science, Peking University, Beijing, China
- * E-mail: (QC); (JY)
| | - Jichun Yang
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Beijing, China
- MOE Key Laboratory of Molecular Cardiovascular Science, Peking University, Beijing, China
- * E-mail: (QC); (JY)
| |
Collapse
|
20
|
Adenosine 2A receptor antagonist prevented and reversed liver fibrosis in a mouse model of ethanol-exacerbated liver fibrosis. PLoS One 2013; 8:e69114. [PMID: 23874883 PMCID: PMC3715448 DOI: 10.1371/journal.pone.0069114] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 06/06/2013] [Indexed: 12/26/2022] Open
Abstract
UNLABELLED The effect of moderate alcohol consumption on liver fibrosis is not well understood, but evidence suggests that adenosine may play a role in mediating the effects of moderate ethanol on tissue injury. Ethanol increases the concentration of adenosine in the liver. Adenosine 2A receptor (A2AR) activation is known to enhance hepatic stellate cell (HSC) activation and A2AR deficient mice are protected from fibrosis in mice. Making use of a novel mouse model of moderate ethanol consumption in which female C57BL/6J mice were allowed continued access to 2% (vol/vol) ethanol (11% calories) or pair-fed control diets for 2 days, 2 weeks or 5 weeks and superimposed with exposure to CCl4, we tested the hypothesis that moderate ethanol consumption increases fibrosis in response to carbon tetrachloride (CCl4) and that treatment of mice with an A2AR antagonist prevents and/or reverses this ethanol-induced increase in liver fibrosis. Neither the expression or activity of CYP2E1, required for bio-activation of CCl4, nor AST and ALT activity in the plasma were affected by ethanol, indicating that moderate ethanol did not increase the direct hepatotoxicity of CCl4. However, ethanol feeding enhanced HSC activation and exacerbated liver fibrosis upon exposure to CCl4. This was associated with an increased sinusoidal angiogenic response in the liver. Treatment with A2AR antagonist both prevented and reversed the ability of ethanol to exacerbate liver fibrosis. CONCLUSION Moderate ethanol consumption exacerbates hepatic fibrosis upon exposure to CCl4. A2AR antagonism may be a potential pharmaceutical intervention to decrease hepatic fibrosis in response to ethanol.
Collapse
|
21
|
Ahmad A, Schaack JB, White CW, Ahmad S. Adenosine A2A receptor-dependent proliferation of pulmonary endothelial cells is mediated through calcium mobilization, PI3-kinase and ERK1/2 pathways. Biochem Biophys Res Commun 2013; 434:566-71. [PMID: 23583199 DOI: 10.1016/j.bbrc.2013.03.115] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 03/26/2013] [Indexed: 12/13/2022]
Abstract
Hypoxia and HIF-2α-dependent A2A receptor expression and activation increase proliferation of human lung microvascular endothelial cells (HLMVECs). This study was undertaken to investigate the signaling mechanisms that mediate the proliferative effects of A2A receptor. A2A receptor-mediated proliferation of HLMVECs was inhibited by intracellular calcium chelation, and by specific inhibitors of ERK1/2 and PI3-kinase (PI3K). The adenosine A2A receptor agonist CGS21680 caused intracellular calcium mobilization in controls and, to a greater extent, in A2A receptor-overexpressing HLMVECs. Adenoviral-mediated A2A receptor overexpression as well as receptor activation by CGS21680 caused increased PI3K activity and Akt phosphorylation. Cells overexpressing A2A receptor also manifested enhanced ERK1/2 phosphorylation upon CGS21680 treatment. A2A receptor activation also caused enhanced cAMP production. Likewise, treatment with 8Br-cAMP increased PI3K activity. Hence A2A receptor-mediated cAMP production and PI3K and Akt phosphorylation are potential mediators of the A2A-mediated proliferative response of HLMVECs. Cytosolic calcium mobilization and ERK1/2 phosphorylation are other critical effectors of HLMVEC proliferation and growth. These studies underscore the importance of adenosine A2A receptor in activation of survival and proliferative pathways in pulmonary endothelial cells that are mediated through PI3K/Akt and ERK1/2 pathways.
Collapse
Affiliation(s)
- Aftab Ahmad
- Pediatric Airway Research Center, Department of Pediatrics, Aurora, CO 80045, USA.
| | | | | | | |
Collapse
|
22
|
Imarisio C, Alchera E, Sutti S, Valente G, Boccafoschi F, Albano E, Carini R. Adenosine A(2a) receptor stimulation prevents hepatocyte lipotoxicity and non-alcoholic steatohepatitis (NASH) in rats. Clin Sci (Lond) 2012; 123:323-332. [PMID: 22439844 DOI: 10.1042/cs20110504] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
NEFA (non-esterified 'free' fatty acid)-mediated lipotoxicity plays a critical role in the pathogenesis of NASH (non-alcoholic steatohepatitis). In the light of the growing need for new therapeutic options for NASH, we investigated the action of A2aR (adenosine A(2a) receptor) stimulation against lipotoxicity. The effects of the A(2a)R agonist CGS21680 [2-p-(2-carboxyethyl)phenethylamino-5'-N-ethylcarboxyamidoadenosine] were evaluated 'in vitro' in liver cells exposed to SA (stearic acid) and 'in vivo' in rats with NASH induced by 8 weeks of feeding with an MCD diet (methionine/choline-deficient diet). In cultured hepatocytes, SA promoted apoptosis by inducing MKK4 (mitogen-activated protein kinase kinase 4)/SEK1 (stress-activated protein kinase/extracellular-signal-regulated kinase kinase-1) and JNK-1/2 (c-Jun N-terminal kinase-1/2) activation. CGS21680 addition prevented JNK-1/2 activation and reduced apoptosis without interfering with lipid accumulation. CGS21680 action required PI3K (phosphoinositide 3-kinase)/Akt-mediated block of MKK4/SEK1. Consistently, PI3K inhibition with wortmannin abolished the cytoprotective action of CGS21680 and reverted MKK4 inhibition. SA lipotoxicity was also prevented by transfecting HTC cells with a specific MKK4/SEK1 siRNA (small interfering RNA). In rats receiving the MCD diet, the development of NASH was associated with MKK4/SEK1 and JNK-1/2 activation. CGS21680 (0.5 mg/kg of body weight, intraperitoneal) administration to MCD-fed rats prevented JNK-1/2 activation by acting on MKK4/SEK1. CGS21680 also effectively reduced NASH-associated ALT (alanine aminotransferase) release, hepatocyte apoptosis, liver inflammation and fibrosis without affecting hepatic steatosis. Taken together, these results demonstrate that, by inhibiting JNK-1/2, A(2a)R stimulation reduces lipotoxicity and ameliorates NASH, giving a rationale to investigate A(2a)R agonists as possible new therapeutic agents in preventing fatty liver progression to NASH.
Collapse
Affiliation(s)
- Chiara Imarisio
- Department of Health Sciences, University 'A. Avogadro', Novara 28100, Italy
| | | | | | | | | | | | | |
Collapse
|
23
|
Yang X, Qin L, Liu J, Tian L, Qian H. 17β-Estradiol protects the liver against cold ischemia/reperfusion injury through the Akt kinase pathway. J Surg Res 2012; 178:996-1002. [PMID: 22835949 DOI: 10.1016/j.jss.2012.07.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Revised: 06/22/2012] [Accepted: 07/03/2012] [Indexed: 10/28/2022]
Abstract
BACKGROUND Hepatic ischemia-reperfusion (IR) injury occurs during liver resection and transplantation. Recent studies have shown that 17β-estradiol (E2) can protect the heart and liver against warm IR. The present study focused on the cytoprotective effects of E2 on cold IR injury to the liver. MATERIALS AND METHODS Sprague-Dawley male rats were randomly divided into three groups: sham, IR, and IR plus E2. The model of rat orthotopic liver transplantation was used. The rats in the IR plus E2 group were intraperitoneally injected with E2 (100 μg/kg/d) for 7 d before surgery. The sham and IR group received the same quantity of saline. The donor livers were then orthotopically transplanted into rats after cold ischemia preservation for 4 h at 4°C lactated Ringer's solution. After 6 h reperfusion, liver function, bile flow volume, hepatocyte apoptosis, and activation of Akt, glycogen synthase kinase-3β, and Bcl-2-associated death promoter were assessed. The survival rate of the rats was also investigated. RESULTS The administration of E2 significantly prolonged the survival of liver grafts by improving liver function and decreasing hepatocyte apoptosis. Rats undergoing E2 demonstrated a greater level activation of Akt in the liver compared with the IR group. In addition, E2 also inhibited the activities of glycogen synthase kinase-3β, Bcl-2-associated death promoter, and caspase-3-induced by IR injury. CONCLUSIONS E2 pretreatment attenuated the hepatocellular damage caused by hepatic cold IR injury through the Akt pathway. Estrogen therapy might be important in clinical settings associated with cold IR injury during liver transplantation.
Collapse
Affiliation(s)
- Xiaohua Yang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | | | | | | | | |
Collapse
|
24
|
Wang D, Ma Y, Li Z, Kang K, Sun X, Pan S, Wang J, Pan H, Liu L, Liang D, Jiang H. The role of AKT1 and autophagy in the protective effect of hydrogen sulphide against hepatic ischemia/reperfusion injury in mice. Autophagy 2012; 8:954-962. [PMID: 22694815 PMCID: PMC3427260 DOI: 10.4161/auto.19927] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Hydrogen sulphide (H 2S) exerts a protective effect in hepatic ischemia-reperfusion (I/R) injury. However, the exact mechanism of H 2S action remains largely unknown. This study was designed to investigate the role of the PtdIns3K-AKT1 pathways and autophagy in the protective effect of H 2S against hepatic I/R injury. Primary cultured mouse hepatocytes and livers with or without NaHS (a donor of H 2S) preconditioning were exposed to anoxia/reoxygenation (A/R) and I/R, respectively. In certain groups, they were also pretreated with LY294002 (AKT1-specific inhibitor), 3-methyladenine (3MA, autophagy inhibitor) or rapamycin (autophagy enhancer), alone or simultaneously. Cell viability, expression of P-AKT1, T-AKT1, LC3 and BECN1 were examined. The severity of liver injury was measured by the levels of serum aminotransferase and inflammatory cytokine, apoptosis and histological examination. GFP-LC3 redistribution and transmission electron microscopy were used to test the activity of autophagy. H 2S preconditioning activated PtdIns3K-AKT1 signaling in hepatocytes. LY294002 could abolish the AKT1 activation and attenuate the protective effect of H 2S on hepatocytes A/R and hepatic I/R injuries. H 2S suppressed hepatic autophagy in vitro and in vivo. Further reducing autophagy by 3MA also diminished the protective effect of H 2S, while rapamycin could reverse the autophagy inhibitory effect and enhance the protective effect of H 2S against hepatocytes A/R and hepatic I/R injuries, consequently. Taken together, H 2S protects against hepatocytic A/R and hepatic I/R injuries, at least in part, through AKT1 activation but not autophagy. An autophagy agonist could be applied to potentiate this hepatoprotective effect by reversing the autophagy inhibition of H 2S.
Collapse
Affiliation(s)
- Dawei Wang
- Key Laboratory of Hepatosplenic Surgery; Department of General Surgery; the First Affiliated Hospital of Harbin Medical University; Harbin, China
| | - Yong Ma
- Key Laboratory of Hepatosplenic Surgery; Department of General Surgery; the First Affiliated Hospital of Harbin Medical University; Harbin, China
| | - Zhengtian Li
- Key Laboratory of Hepatosplenic Surgery; Department of General Surgery; the First Affiliated Hospital of Harbin Medical University; Harbin, China
| | - Kai Kang
- Department of Intensive Care Unit; the First Affiliated Hospital of Harbin Medical University; Harbin, China
| | - Xueying Sun
- Key Laboratory of Hepatosplenic Surgery; Department of General Surgery; the First Affiliated Hospital of Harbin Medical University; Harbin, China
- Department of Molecular Medicine and Pathology; Faculty of Medical and Health Sciences; University of Auckland; Auckland, New Zealand
| | - Shangha Pan
- Key Laboratory of Hepatosplenic Surgery; Department of General Surgery; the First Affiliated Hospital of Harbin Medical University; Harbin, China
| | - Jizhou Wang
- Key Laboratory of Hepatosplenic Surgery; Department of General Surgery; the First Affiliated Hospital of Harbin Medical University; Harbin, China
| | - Huayang Pan
- Key Laboratory of Hepatosplenic Surgery; Department of General Surgery; the First Affiliated Hospital of Harbin Medical University; Harbin, China
| | - Lianxin Liu
- Key Laboratory of Hepatosplenic Surgery; Department of General Surgery; the First Affiliated Hospital of Harbin Medical University; Harbin, China
| | - Desen Liang
- Key Laboratory of Hepatosplenic Surgery; Department of General Surgery; the First Affiliated Hospital of Harbin Medical University; Harbin, China
| | - Hongchi Jiang
- Key Laboratory of Hepatosplenic Surgery; Department of General Surgery; the First Affiliated Hospital of Harbin Medical University; Harbin, China
| |
Collapse
|
25
|
Abstract
Melatonin is a potent scavenger of reactive oxygen species and a strong antioxidant. Melatonin exerts protective effects against damage by the enhancing the Akt signal pathway, thus regulating apoptotic cell death. Akt phosphorylates pro-apoptotic proteins such as Bad and FoxO1 and inhibits the pro-apoptotic functions of these proteins. This study investigated the protective effects of melatonin through Akt and its downstream targets, Bad and FoxO1, in hepatic ischemia-reperfusion (I/R) damage. Adult mice were subjected to 1 h of hepatic ischemia and 3 h of reperfusion. Hepatic ischemia was induced by occlusions of the hepatic artery, portal vein, and bile duct. Melatonin (10 mg/kg, i.p.) or vehicle was administrated 15 min prior to ischemia and just before reperfusion. Serum aspartate aminotransferase and alanine aminotransferase levels were higher in I/R group than in sham-operated group. Melatonin attenuated increases in these levels. Moreover, melatonin attenuates injury-induced increases in positive TUNEL staining in hepatic tissues. Hepatic I/R injury induced reductions in the Akt up-stream target, PDK1 phosphorylation. The levels of phospho-Akt, phospho-Bad, and phospho-FoxO1 were decreased in vehicle-treated animals. However, melatonin prevented hepatic I/R injury-induced decreases in these proteins levels. Moreover, the interaction levels between phospho-Bad and 14-3-3 and between phospho-FoxO1 and 14-3-3 are reduced in vehicle-treated animals, and melatonin attenuated decreases in the binding levels of these proteins. 14-3-3 exerts an anti-apoptotic function by sequestration of Bad and FoxO1. These findings suggest that melatonin exerts protective effects in case of hepatic I/R damage by maintaining the binding of phospho-Bad and 14-3-3 and the binding of phospho-FoxO1 and 14-3-3, thus preventing activation of apoptotic cell death.
Collapse
Affiliation(s)
- Phil-Ok Koh
- Department of Anatomy, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea.
| |
Collapse
|
26
|
Abstract
Obesity is a global epidemic with more than 1 billion overweight adults and at least 300 million obese patients worldwide. Diabetes is characterized by a defect in insulin secretion or a decrease in sensitivity to insulin, which results in elevated fasting blood glucose. Both obesity and elevated fasting glucose are risk factors for nonalcoholic fatty liver disease, a disease spectrum that includes hepatic steatosis (nonalcoholic fatty liver), nonalcoholic steatohepatitis (NASH), fibrosis, and cirrhosis. Increased adiposity and insulin resistance contribute to the progression from NASH to fibrosis through the development of a profibrotic mileau in the liver, including increased hepatocellular death, increased reactive oxygen species generation, and an altered adipokine/cytokine balance. This review will summarize recent advances in our understanding of the pathological interactions among excessive fat accumulation, insulin resistance, and hepatic fibrogenesis and discuss specific molecular pathways that may be of interest in the development of therapeutic interventions to prevent and/or reverse hepatic fibrosis.
Collapse
Affiliation(s)
- Dian J Chiang
- Dept. of Gastroenterology and Hepatology, Cleveland Clinic, OH 44195, USA.
| | | | | |
Collapse
|
27
|
Dal Ponte C, Alchera E, Follenzi A, Imarisio C, Prat M, Albano E, Carini R. Pharmacological postconditioning protects against hepatic ischemia/reperfusion injury. Liver Transpl 2011; 17:474-82. [PMID: 21445931 DOI: 10.1002/lt.22256] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Postconditioning is a procedure based on the induction of intracellular protective reactions immediately after the onset of reperfusion. Because of the growing need to prevent ischemia/reperfusion (I/R) injury during liver surgery and transplantation, we investigated the possibility of pharmacologically inducing hepatic postconditioning. The effects of the adenosine A2A receptor agonist 2p-(2-carboxyethyl)-phenyl-amino-5'-N-ethylcarboxyamido-adenosine (CGS21680; 5 μmol/L) and the phosphatase and tensin homologue deleted from chromosome 10 (PTEN) inhibitor dipotassium bisperoxo-(5-hydroxypyridine-2-carboxyl)-oxovanadate [bpV(HOpic); 250 nmol/L] were investigated in primary rat hepatocytes during reoxygenation after 24 hours of cold storage and in an in vivo model of rat liver warm I/R. The addition of CGS21680 at reoxygenation significantly reduced hepatocyte death through the activation of the phosphoinositide 3-kinase (PI3K)-protein kinase B (PKB)/Akt signal pathway and through the reduction of the intracellular level of PTEN. PTEN lowering was associated with the increased generation of reactive oxygen species after A2A receptor-mediated stimulation of β-nicotinamide adenine dinucleotide phosphate oxidase (NOX). The inhibition of PI3K or NOX with wortmannin or diphenyleneiodonium chloride, respectively, and the addition of the antioxidant N,N'-diphenyl-p-phenylenediamine reversed the effects of CGS21680. The PTEN inhibitor bpV(HOpic) mimicked the protection provided by CGS21680 against reoxygenation damage. An in vivo rat treatment with CGS21680 or bpV(HOpic) during reperfusion after 1 hour of partial hepatic ischemia also promoted PKB/Akt activation and ameliorated alanine aminotransferase release and histological lesions induced by 2 hours of reperfusion. We conclude that adenosine A2A receptor agonists and PTEN inhibitors are possibly useful agents for the pharmacological induction of postconditioning in the liver.
Collapse
Affiliation(s)
- Caterina Dal Ponte
- Department of Medical Sciences, Amedeo Avogadro University of East Piedmont, Novara, Italy
| | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Ischemia/reperfusion (I/R) injury still represents an important cause of morbidity following hepatic surgery and limits the use of marginal livers in hepatic transplantation. Transient blood flow interruption followed by reperfusion protects tissues against damage induced by subsequent I/R. This process known as ischemic preconditioning (IP) depends upon intrinsic cytoprotective systems whose activation can inhibit the progression of irreversible tissue damage. Compared to other organs, liver IP has additional features as it reduces inflammation and promotes hepatic regeneration. Our present understanding of the molecular mechanisms involved in liver IP is still largely incomplete. Experimental studies have shown that the protective effects of liver IP are triggered by the release of adenosine and nitric oxide and the subsequent activation of signal networks involving protein kinases such as phosphatidylinositol 3-kinase, protein kinase C δ/ε and p38 MAP kinase, and transcription factors such as signal transducer and activator of transcription 3, nuclear factor-κB and hypoxia-inducible factor 1. This article offers an overview of the molecular events underlying the preconditioning effects in the liver and points to the possibility of developing pharmacological approaches aimed at activating the intrinsic protective systems in patients undergoing liver surgery.
Collapse
|
29
|
Huber-Ruano I, Pinilla-Macua I, Torres G, Casado FJ, Pastor-Anglada M. Link between high-affinity adenosine concentrative nucleoside transporter-2 (CNT2) and energy metabolism in intestinal and liver parenchymal cells. J Cell Physiol 2010; 225:620-30. [PMID: 20506327 DOI: 10.1002/jcp.22254] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Concentrative nucleoside transporter 2 (CNT2) is a high-affinity adenosine transporter that may play physiological roles beyond nucleoside salvage. Previous reports relate CNT2 function to modulation of purinergic signaling and energy metabolism in intestinal and liver parenchymal cells (Duflot et al., 2004, Mol Cell Biol 24:2710-2719; Aymerich et al., 2006, J Cell Sci 119:1612-1621). In the present study, to further examine the link between CNT2 and energy metabolism, CNT2 protein partners were identified using the bacterial two-hybrid and GST pull-down approaches. The N-terminal segment of CNT2 was used as bait, since proteins lacking this domain display impaired plasma membrane insertion and intracellular retention. Glucose-regulated protein 58 (GRP58) was identified as a potential rCNT2 partner in pull-down experiments. Two-hybrid screening performed against a liver human cDNA library led to the identification of aldolase B as another hCNT2 partner. Aldolase B-RFP and endogenous GRP58 separately co-localized with CNT2 in HeLa cells transfected with YFPrCNT2. CNT2 interaction with GRP58 was validated using co-immunoprecipitation experiments. In HeLa cells, fluorescence resonance energy transfer (FRET) efficiency increased upon fructose addition, consistent with a transient interaction between aldolase B and the transporter. The physiological basis for in vivo interactions was derived from experiments in which GRP58 was inhibited or overexpressed and aldolase B activity stimulated towards glycolysis. GRP58 appeared to be a negative effector of CNT2 function, whereas aldolase B flux modulated CNT2 activity via a mechanism involving acquisition of higher affinity for its substrates. These findings support the theory that CNT2 plays roles other than salvage and establishes links with energy metabolism.
Collapse
Affiliation(s)
- Isabel Huber-Ruano
- Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina, Universitat de Barcelona and CIBER EHD, Barcelona, Spain
| | | | | | | | | |
Collapse
|
30
|
Negative regulation of diacylglycerol kinase theta mediates adenosine-dependent hepatocyte preconditioning. Cell Death Differ 2010; 17:1059-68. [PMID: 20057501 DOI: 10.1038/cdd.2009.210] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In liver ischemic preconditioning (IP), stimulation of adenosine A2a receptors (A2aR) prevents ischemia/reperfusion injury by promoting diacylglycerol-mediated activation of protein kinase C (PKC). By concerting diacylglycerol to phosphatidic acid, diacylglycerol kinases (DGKs) act as terminator of diacylglycerol signalling. This study investigates the role of DGK in the development of hepatocyte IP. DGK activity and cell viability were evaluated in isolated rat hepatocytes preconditioned by 10 min hypoxia followed by 10 min re-oxygenation or by the treatment with the A2aR agonist, CGS21680, and subsequently exposed to prolonged hypoxia. We observed that after IP or A2aR activation, a decrease in DGK activity was associated with the onset of hepatocyte tolerance to hypoxia. CGS21680-induced stimulation of A2aR specifically inhibited DGK isoform theta by activating RhoA-GTPase. Consistently, both siRNA-mediated downregulation of DGK theta and hepatocyte pretreatment with the DGK inhibitor R59949 induced cell tolerance to hypoxia. The pharmacological inhibition of DGK was associated with the diacylglycerol-dependent activation of PKC delta and epsilon and of their downstream target p38 MAPK. In conclusion, we unveil a novel signalling pathway contributing to the onset of hepatocyte preconditioning, which through RhoA-GTPase, couples A2aR to the downregulation of DGK. Such an inhibition is essential for the sustained accumulation of diacylglycerol required for triggering PKC-mediated survival signals.
Collapse
|
31
|
Role of ischemic preconditioning in liver surgery and hepatic transplantation. J Gastrointest Surg 2009; 13:2074-83. [PMID: 19404711 DOI: 10.1007/s11605-009-0878-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2009] [Accepted: 03/24/2009] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The purpose of this review is to summarize intraoperative surgical strategies available to decrease ischemia-reperfusion injury associated with liver resection and liver transplantation. MATERIAL AND METHOD We conducted a critical review of the literature evaluating the potential applications of hepatic ischemic preconditioning (IPC) for hepatic resection surgery and liver transplantation. In addition, we provide a basic bench-to-bedside summary of the liver physiology and cell signaling mechanisms that account for the protective effects seen with hepatic IPC.
Collapse
|
32
|
Richmond SR, Touchberry CD, Gallagher PM. Forskolin attenuates the action of insulin on the Akt–mTOR pathway in human skeletal muscle. Appl Physiol Nutr Metab 2009; 34:916-25. [DOI: 10.1139/h09-096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Forskolin (FSK) is capable of both stimulating and inhibiting the intracellular signaling pathways of protein synthesis tissues other than skeletal muscle. The purpose of this investigation was to determine if FSK administration affects various elements of the protein kinase B (Akt)–mammalian target of rapamycin (mTOR) pathway in human skeletal muscle. Ten (n = 10) healthy, young (21.6 ± 1.3 years), nonobese (body mass index = 25.5 ± 3.5 kg·m–2), recreationally active males were selected for participation. Following an 8 h fast, 2 muscle biopsies of the vastus lateralis were performed. The samples were sectioned and exposed to 4 in vitro treatment conditions: basal, FSK, insulin (INS), and FSK+INS. The samples were then analyzed for total and phosphorylated levels of Akt, mTOR, S6 kinase (S6K1), and 4E binding protein (4EBP1). Akt phosphorylation was significantly greater in the INS-treated samples compared with the basal and FSK conditions (p = 0.007). Furthermore, the ratio of phosphorylated Akt to total Akt (P/T) was higher in the INS samples compared with the basal and FSK samples (p = 0.001). There were no differences in mTOR phosphorylation among the 4 groups; however, total mTOR was significantly greater in the FSK+INS group (p = 0.006). There were also no differences in phosphorylated or total levels of S6K1 among the 4 groups. However, 4EBP1 phosphorylation was significantly greater in the INS-treated samples compared with the basal (p = 0.003) and FSK (p = 0.004) treatments. There were no differences in the ratio of phosphorylated 4EBP1 to total 4EBP1 (P/T) among the 4 groups. These results indicate that FSK does not activate the Akt–mTOR pathway in human skeletal muscle; however, these results suggest that FSK may inhibit the actions of INS on this pathway.
Collapse
Affiliation(s)
- Scott R. Richmond
- Applied Physiology Laboratory, University of Kansas, Lawrence, KS 66045, USA
| | - Chad D. Touchberry
- Applied Physiology Laboratory, University of Kansas, Lawrence, KS 66045, USA
| | - Philip M. Gallagher
- Applied Physiology Laboratory, University of Kansas, Lawrence, KS 66045, USA
| |
Collapse
|
33
|
Toledo-Pereyra LH, Lopez-Neblina F, Toledo AH. Protein Kinases in Organ Ischemia and Reperfusion. J INVEST SURG 2009; 21:215-26. [DOI: 10.1080/08941930802130149] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
34
|
Variable activation of phosphoinositide 3-kinase influences the response of liver grafts to ischemic preconditioning. J Hepatol 2009; 50:937-47. [PMID: 19303157 DOI: 10.1016/j.jhep.2008.11.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Revised: 11/07/2008] [Accepted: 11/25/2008] [Indexed: 02/08/2023]
Abstract
BACKGROUND/AIMS The efficacy of ischemic preconditioning (IPC) in preventing reperfusion injury in human liver transplants is still questioned. Phosphoinositide-3-kinase (PI3K) is essential for IPC development in rodent livers. This work investigates whether PI3K-dependent signals might account for the inconsistent responses to IPC of transplanted human livers. METHODS Forty livers from deceased donors were randomized to receive or not IPC before recovery. PI3K activation was evaluated in biopsies obtained immediately before IPC and 2 h after reperfusion by measuring the phosphorylation of the PI3K downstream kinase PKB/Akt and the levels of the PI3K antagonist phosphatase tensin-homologue deleted from chromosome 10 (PTEN). RESULTS IPC increased PKB/Akt phosphorylation (p = 0.01) and decreased PTEN levels (p = 0.03) in grafts, but did not significantly ameliorate post-transplant reperfusion injury. By calculating T(2h)/T(0) PKB/Akt phosphorylation ratios, 10/19 (53%) of the preconditioned grafts had ratios above the control threshold (IPC-responsive), while the remaining nine grafts showed ratios comparable to controls (IPC-non-responsive). T(2h)/T(0) PTEN ratios were also decreased (p < or = 0.03) only in IPC-responsive grafts. The patients receiving IPC-responsive organs had ameliorated (p < or = 0.05) post-transplant aminotransferase and bilirubin levels, while prothrombin activity was unchanged. CONCLUSIONS Impaired PI3K signaling might account for the variability in the responses to IPC of human grafts from deceased donors.
Collapse
|
35
|
Gates A, Hohenester S, Anwer MS, Webster CRL. cAMP-GEF cytoprotection by Src tyrosine kinase activation of phosphoinositide-3-kinase p110 beta/alpha in rat hepatocytes. Am J Physiol Gastrointest Liver Physiol 2009; 296:G764-74. [PMID: 19196950 PMCID: PMC2670669 DOI: 10.1152/ajpgi.90622.2008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Cyclic AMP protects against hepatocyte apoptosis by a protein kinase A-independent cAMP-GEF/phosphoinositide-3-kinase (PI3K)/Akt signaling pathway. However, the signaling pathway coupling cAMP-GEF with PI3K is unknown. The aim of this study was to investigate the role of Src tyrosine kinases (Src-TYK) and PI3K-p110 isoforms in this pathway. Studies were done in rat hepatocytes using the hydrophobic bile acid glycochenodeoxycholic acid (GCDC) to induce apoptosis. cAMP-binding guanine nucleotide exchange factors (cAMP-GEFs) were selectively activated by using 4-(4-chloro-phenylthio)-2'-O-methyladenosine-3'-5'-cyclic monophosphate (CPT-2-Me-cAMP), which sequentially phosphorylated Src-TYK (within 1 min) followed by Akt (within 5 min). The Src inhibitors PP2 and SU6656 inhibited basal and CPT-2-Me-cAMP-mediated Src and Akt phosphorylation. These inhibitors had no effect on CPT-2-Me-cAMP-mediated activation of Rap GTPases. CPT-2-Me-cAMP induced transient Src dependent autophosphorylation of the epidermal growth factor receptor (EGFR). Inhibition of the EGFR with AG 1478 partially inhibited the ability of CPT-2-Me to phosphorylate Akt. Whereas PP2 completely abolished the protective effect of CPT-2-Me-cAMP in GCDC induced apoptosis, AG 1478 partially inhibited the cytoprotective effect. CPT-2-Me-cAMP treatment resulted in Src-dependent activation of the p110 beta and alpha subunits of PI3K, but only the latter was sensitive to inhibition with AG 1478. In conclusion, activation of cAMP-GEFs results in phosphorylation of Src-TYK and Akt and activation of the p110 beta/alpha subunits of PI3K. Maximal cAMP-GEF-mediated Akt phosphorylation as well as protection from bile acid-induced apoptosis requires activation of Src-TYK and the EGFR. These studies support the existence of two pathways: cAMP-GEF/Rap/Src/PI3Kbeta/Akt and cAMP-GEF/Rap/Src/EGFR/PI3Kalpha/Akt, both of which are necessary for maximal cytoprotective effect of cAMP-GEFs in hepatocytes.
Collapse
Affiliation(s)
- Anna Gates
- Departments of Clinical Sciences and Biomedical Sciences, Tufts Cummings School of Veterinary Medicine, North Grafton, Massachusetts
| | - Simon Hohenester
- Departments of Clinical Sciences and Biomedical Sciences, Tufts Cummings School of Veterinary Medicine, North Grafton, Massachusetts
| | - M. Sawkat Anwer
- Departments of Clinical Sciences and Biomedical Sciences, Tufts Cummings School of Veterinary Medicine, North Grafton, Massachusetts
| | - Cynthia R. L. Webster
- Departments of Clinical Sciences and Biomedical Sciences, Tufts Cummings School of Veterinary Medicine, North Grafton, Massachusetts
| |
Collapse
|
36
|
Abstract
BACKGROUND The negative effects of increased donor age on liver transplantation became evident in deceased donor liver transplantation. In living donor liver transplantation (LDLT), the details remain unclear. METHODS Initially, 137 adult LDLT recipients from August 1996 to May 2005 were divided into two groups (donors <50 years of age: n=99, donors >or= 50 years of age: n=38) for the retrospective study. Then, 24 recipients who received LDLT from June 2005 to July 2006 were divided into two groups: group 1 (donors <50 years of age, n=14) and group 2 (donors >or= 50 years of age, n=10) and enrolled in the prospective study to analyze their clinical course and prognostic factors in the aged graft. RESULTS In the retrospective study, the younger donor group had significantly better survival than that of the aged donor group (P=0.015, Log rank test). In the prospective study, the postoperative graft functions showed that the serum total bilirubin levels were significantly lower in group 1 (P<0.02, by ANOVA analysis). The phosphorylated-Signal Transducer and Activator of Transcription3 expression at 4 hr after reperfusion (RT2) in group 2 was significantly lower than that in group 1. At RT2, the expressions were up-regulated in group 1, but were down-regulated in group 2. The serum 8-hydroxydeoxyguanosine value became significantly higher in group 1 two weeks after LDLT. CONCLUSIONS In the near term, Signal Transducer and Activator of Transcription3 gene induction during cold preservation may be of great use in improving the outcome of aged grafts in LDLT.
Collapse
|
37
|
Alchera E, Tacchini L, Imarisio C, Dal Ponte C, De Ponti C, Gammella E, Cairo G, Albano E, Carini R. Adenosine-dependent activation of hypoxia-inducible factor-1 induces late preconditioning in liver cells. Hepatology 2008; 48:230-9. [PMID: 18506850 DOI: 10.1002/hep.22249] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
UNLABELLED The cellular mechanisms by which ischemic preconditioning increases liver tolerance to ischemia/reperfusion injury are still poorly understood. This study investigated the role of the hypoxia-inducible factor-1 (HIF-1) in the protection associated with the late phase of liver preconditioning. Late preconditioning was induced in primary cultured rat hepatocytes by a transient (10 minute) hypoxic stress or by 15 minutes incubation with the adenosine A(2A) receptors agonist CGS21680 24 hours before exposure to 90 minutes of hypoxia in a serum-free medium. Late preconditioning induced the nuclear translocation of HIF-1 and the expression of carbonic anhydrase IX (CAIX), a HIF-1-regulated transmembrane enzyme that catalyzes bicarbonate production. Such effects were associated with prevention of hepatocyte killing by hypoxia and the amelioration of intracellular acidosis and Na+ accumulation. The inhibition of PKC-mediated and PI3-kinase-mediated signals with, respectively, chelerythrine and wortmannin abolished HIF-1 activation and blocked both CAIX expression and the protective action of late preconditioning. CAIX expression was also prevented by interfering with the transcriptional activity of HIF-1 using a dominant negative HIF-1beta subunit. The inhibition of CAIX with acetazolamide or the block of bicarbonate influx with disodium-4-acetamido-4'-isothiocyanato-stilben-2,2'-disulfonate also reverted the protective effects of late preconditioning on intracellular acidosis and Na+ accumulation. CONCLUSION The stimulation of adenosine A(2A) receptors induced late preconditioning in liver cells through the activation of HIF-1. HIF-1-induced expression of CAIX increases hepatocyte tolerance to ischemia by maintaining intracellular Na+ homeostasis. These observations along with the importance of HIF-1 in regulating cell survival indicates HIF-1 activation as a possible key event in liver protection by late preconditioning.
Collapse
Affiliation(s)
- Elisa Alchera
- Dipartimento di Scienze Mediche, Università "A. Avogadro", Novara, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Adenosine regulates the function of the innate and adaptive immune systems through targeting virtually every cell type that is involved in orchestrating an immune/inflammatory response. Of the four adenosine receptors (A(1), A(2A), A(2B), A(3)), A(2A) receptors have taken center stage as the primary anti-inflammatory effectors of extracellular adenosine. This broad, anti-inflammatory effect of A(2A) receptor activation is a result of the predominant expression of A(2A) receptors on monocytes/macrophages, dendritic cells, mast cells, neutrophils, endothelial cells, eosinophils, epithelial cells, as well as lymphocytes, NK cells, and NKT cells. A(2A) receptor activation inhibits early and late events occurring during an immune response, which include antigen presentation, costimulation, immune cell trafficking, immune cell proliferation, proinflammatory cytokine production, and cytotoxicity. In addition to limiting inflammation, A(2A) receptors participate in tissue remodeling and reparation. Consistent with their multifaceted, immunoregulatory action on immune cells, A(2A) receptors have been shown to impact the course of a wide spectrum of ischemic, autoimmune, infectious, and allergic diseases. Here, we review the regulatory roles of A(2A) receptors in immune/inflammatory diseases of various organs, including heart, lung, gut, liver, kidney, joints, and brain, as well as the role of A(2A) receptors in regulating multiple organ failure and sepsis.
Collapse
Affiliation(s)
- György Haskó
- UMDNJ-New Jersey Medical School, 185 South Orange Avenue, University Heights, Newark, NJ 07103, USA.
| | | |
Collapse
|
39
|
Kuboki S, Shin T, Huber N, Eismann T, Galloway E, Schuster R, Blanchard J, Zingarelli B, Lentsch AB. Peroxisome proliferator-activated receptor-gamma protects against hepatic ischemia/reperfusion injury in mice. Hepatology 2008; 47:215-24. [PMID: 18085707 DOI: 10.1002/hep.21963] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
UNLABELLED The function of peroxisome proliferator-activated receptor-gamma (PPARgamma) in hepatic inflammation and injury is unclear. In this study, we sought to determine the role of PPARgamma in hepatic ischemia/reperfusion injury in mice. Male mice were subjected to 90 minutes of partial hepatic ischemia followed by up to 8 hours of reperfusion. PPARgamma was found to be constitutively activated in hepatocytes but not in nonparenchymal cells. Upon induction of ischemia, hepatic PPARgamma activation rapidly decreased and remained suppressed throughout the 8-hour reperfusion period. This reduced activation was not a result of decreased protein availability as hepatic nuclear PPARgamma, retinoid X receptor-alpha (RXRalpha), and PPARgamma/RXRalpha heterodimer expression was maintained. Accompanying the decrease in PPARgamma activation was a decrease in the expression of the natural ligand 15-deoxy-Delta(12,14)-prostaglandin J(2). This was associated with reduced interaction of PPARgamma and the coactivator, p300. To determine whether PPARgamma activation is hepatoprotective during hepatic ischemia/reperfusion injury, mice were treated with the PPARgamma agonists, rosiglitazone and connecting peptide. These treatments increased PPARgamma activation and reduced liver injury compared to untreated mice. Furthermore, PPARgamma-deficient mice had more liver injury after ischemia/reperfusion than their wild-type counterparts. CONCLUSION These data suggest that PPARgamma is an important endogenous regulator of, and potential therapeutic target for, ischemic liver injury.
Collapse
Affiliation(s)
- Satoshi Kuboki
- Laboratory of Trauma, Sepsis & Inflammation Research, Department of Surgery, University of Cincinnati, Cincinnati, OH 45267-0558, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Zhou D, Xue J, Chen J, Morcillo P, Lambert JD, White KP, Haddad GG. Experimental selection for Drosophila survival in extremely low O(2) environment. PLoS One 2007; 2:e490. [PMID: 17534440 PMCID: PMC1871610 DOI: 10.1371/journal.pone.0000490] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2007] [Accepted: 05/11/2007] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Cellular hypoxia, if severe enough, results usually in injury or cell death. Our research in this area has focused on the molecular mechanisms underlying hypoxic tissue injury to explore strategies to prevent injury or enhance tolerance. The current experiments were designed to determine the genetic basis for adaptation to long term low O(2) environments. METHODOLOGY/PRINCIPAL FINDINGS With long term experimental selection over many generations, we obtained a Drosophila melanogaster strain that can live perpetually in extremely low, normally lethal, O(2) condition (as low as 4% O(2)). This strain shows a dramatic phenotypic divergence from controls, including a decreased recovery time from anoxic stupor, a higher rate of O(2 )consumption in hypoxic conditions, and a decreased body size and mass due to decreased cell number and size. Expression arrays showed that about 4% of the Drosophila genome altered in expression and about half of the alteration was down-regulation. The contribution of some altered transcripts to hypoxia tolerance was examined by testing the survival of available corresponding P-element insertions (and their excisions) under extremely low O(2) conditions. We found that down-regulation of several candidate genes including Best1, broad, CG7102, dunce, lin19-like and sec6 conferred severe hypoxia tolerance in Drosophila. CONCLUSIONS/SIGNIFICANCE We have identified a number of genes that play an important role in the survival of a selected Drosophila strain in extremely low O(2) conditions, selected by decreasing O(2) availability over many generations. Because of conservation of pathways, we believe that such genes are critical in hypoxia adaptation in physiological or pathological conditions not only in Drosophila but also in mammals.
Collapse
Affiliation(s)
- Dan Zhou
- Departments of Pediatrics, Section of Respiratory Medicine, and Neuroscience, University of California San Diego, La Jolla, California, United States of America
- The Rady Children's Hospital - San Diego, San Diego, California, United States of America
| | - Jin Xue
- Departments of Pediatrics, Section of Respiratory Medicine, and Neuroscience, University of California San Diego, La Jolla, California, United States of America
- The Rady Children's Hospital - San Diego, San Diego, California, United States of America
| | - Jianming Chen
- Department of Immunology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Patrick Morcillo
- Department of Cell Biology, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York, United States of America
| | - J. David Lambert
- Department of Biology, University of Rochester, Rochester, New York, United States of America
| | - Kevin P. White
- Institute for Genomics & Systems Biology and Departments of Human Genetics and Ecology and Evolution, The University of Chicago, Chicago, Illinois, United States of America
- * To whom correspondence should be addressed. E-mail: (GH), (KP)
| | - Gabriel G. Haddad
- Departments of Pediatrics, Section of Respiratory Medicine, and Neuroscience, University of California San Diego, La Jolla, California, United States of America
- The Rady Children's Hospital - San Diego, San Diego, California, United States of America
- * To whom correspondence should be addressed. E-mail: (GH), (KP)
| |
Collapse
|
41
|
De Ponti C, Carini R, Alchera E, Nitti MP, Locati M, Albano E, Cairo G, Tacchini L. Adenosine A2areceptor-mediated, normoxic induction of HIF-1 through PKC and PI-3K-dependent pathways in macrophages. J Leukoc Biol 2007; 82:392-402. [PMID: 17505024 DOI: 10.1189/jlb.0107060] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Adenosine released by cells in injurious or hypoxic environments has tissue-protecting and anti-inflammatory effects, which are also a result of modulation of macrophage functions, such as vascular endothelial growth factor (VEGF) production. As VEGF is a well-known target of hypoxia-inducible factor 1 (HIF-1), we hypothesized that adenosine may activate HIF-1 directly. Our studies using subtype-specific adenosine receptor agonists and antagonists showed that by activating the A(2A) receptor, adenosine treatment induced HIF-1 DNA-binding activity, nuclear accumulation, and transactivation capacity in J774A.1 mouse macrophages. Increased HIF-1 levels were also found in adenosine-treated mouse peritoneal macrophages. The HIF-1 activation induced by the A(2A) receptor-specific agonist CGS21680 required the PI-3K and protein kinase C pathways but was not mediated by changes in iron levels. Investigation of the molecular basis of HIF-1 activation revealed the involvement of transcriptional and to a larger extent, translational mechanisms. HIF-1 induction triggered the expression of HIF-1 target genes involved in cell survival (aldolase, phosphoglycerate kinase) and VEGF but did not induce inflammation-related genes regulated by HIF-1, such as TNF-alpha or CXCR4. Our results show that the formation of adenosine and induction of HIF-1, two events which occur in response to hypoxia, are linked directly and suggest that HIF-1 activation through A(2A) receptors may contribute to the anti-inflammatory and tissue-protecting activity of adenosine.
Collapse
|
42
|
Hsieh YC, Yu HP, Frink M, Suzuki T, Choudhry MA, Schwacha MG, Chaudry IH. G protein-coupled receptor 30-dependent protein kinase A pathway is critical in nongenomic effects of estrogen in attenuating liver injury after trauma-hemorrhage. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 170:1210-8. [PMID: 17392161 PMCID: PMC1829455 DOI: 10.2353/ajpath.2007.060883] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Although nongenomic effects of 17beta-estradiol (E2) are mediated via the estrogen receptor alpha (ER-alpha), the existence of another novel ER, G protein-coupled receptor 30 (GPR30), has been suggested as a candidate for triggering a broad range of E2-mediated signaling. GPR30 also acts independently of the ER to promote activation of the protein kinase A (PKA) pathway, which protects cells from apoptosis through Bcl-2. In this study, we examined whether the salutary effects of E2 in attenuating hepatic injury after trauma-hemorrhage are mediated via GPR30- or ER-alpha-regulated activation of PKA-dependent signaling. At 2 hours after trauma-hemorrhage, administration of E2-conjugated to bovine serum albumin (E2-BSA, membrane impermeable) or E2 induced the up-regulation of ER-alpha and GPR30 and attenuated hepatic injury. This was accompanied by increases in PKA activity and Bcl-2 expression. Inhibition of PKA in E2-BSA-treated trauma-hemorrhage rats by PKA inhibitor H89 prevented the E2-BSA attenuation of hepatic injury. Isolated hepatocytes were transfected with small interfering RNA to suppress GPR30 or ER. We found that suppression of GPR30 but not ER-alpha prevented E2-BSA- or E2-induced PKA activation and Bcl-2 expression. These results suggest that the nongenomic salutary effect of E2 in reducing hepatic injury after trauma-hemorrhage is mediated through the PKA-dependent pathway via GPR30 but not ER-alpha.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Estradiol/administration & dosage
- Estradiol/pharmacology
- Estradiol/therapeutic use
- Estrogen Receptor alpha/genetics
- Estrogen Receptor alpha/metabolism
- Estrogens, Conjugated (USP)/administration & dosage
- Estrogens, Conjugated (USP)/pharmacology
- Estrogens, Conjugated (USP)/therapeutic use
- Glutathione Transferase/blood
- Hemorrhage/blood
- Hemorrhage/etiology
- Hemorrhage/prevention & control
- Hepatocytes/drug effects
- Hepatocytes/metabolism
- Isoenzymes/blood
- Isoquinolines/administration & dosage
- Isoquinolines/pharmacology
- Liver/drug effects
- Liver/injuries
- Liver/metabolism
- Male
- Models, Biological
- Protein Kinase Inhibitors/pharmacology
- Proto-Oncogene Proteins c-bcl-2/metabolism
- RNA, Small Interfering/genetics
- Rats
- Rats, Sprague-Dawley
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/physiology
- Serum Albumin, Bovine/administration & dosage
- Serum Albumin, Bovine/pharmacology
- Serum Albumin, Bovine/therapeutic use
- Signal Transduction/drug effects
- Sulfonamides/administration & dosage
- Sulfonamides/pharmacology
- Transfection
- Wounds and Injuries/complications
Collapse
Affiliation(s)
- Ya-Ching Hsieh
- Center for Surgical Research, University of Alabama at Birmingham, 1670 University Blvd., Volker Hall, Room G094, Birmingham, AL 35294-0019, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Zhang Y, Park TS, Gidday JM. Hypoxic preconditioning protects human brain endothelium from ischemic apoptosis by Akt-dependent survivin activation. Am J Physiol Heart Circ Physiol 2007; 292:H2573-81. [PMID: 17337592 DOI: 10.1152/ajpheart.01098.2006] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Preconditioning-induced ischemic tolerance is well documented in the brain, but cell-specific responses and mechanisms require further elucidation. The aim of this study was to develop an in vitro model of ischemic tolerance in human brain microvascular endothelial cells (HBMECs) and to examine the roles of phosphatidylinositol 3-kinase (PI3-kinase)/Akt and the inhibitor-of- apoptosis protein, survivin, in the ability of hypoxic preconditioning (HP) to protect endothelium from apoptotic cell death. Cultured HBMECs were subjected to HP, followed 16 h later by complete oxygen and glucose deprivation (OGD) for 8 h; cell viability was quantified at 20 h of reoxygenation (RO) by the 3-(4,5-dimethylthiazol)-2,5-diphenyltetrazolium bromide assay. HBMECs were examined at various times after HP or OGD/RO using immunoblotting and confocal laser scanning immunofluorescence microscopy for appearance of apoptotic markers and expression of phosphorylated (p)-Akt and p-survivin. Causal evidence for the participation of the PI3-kinase/Akt pathway in HP-induced protection and p-survivin upregulation was assessed by the PI3-kinase inhibitor LY-294002. HP significantly reduced OGD/RO-induced injury by 50% and also significantly reduced the OGD-induced translocation of apoptosis-inducing factor (AIF) from mitochondria to nucleus and the concomitant cleavage of poly(ADP-ribose) polymerase-1 (PARP-1). PI3-kinase inhibition blocked HP-induced increases in Akt phosphorylation, reversed the effects of HP on OGD-induced AIF translocation and PARP-1 cleavage, blocked HP-induced survivin phosphorylation, and ultimately attenuated HP-induced protection of HBMECs from OGD. Thus HP promotes an antiapoptotic phenotype in HBMECs, in part by activating survivin via the PI3-kinase/Akt pathway. Survivin and other phosphorylation products of p-Akt may be therapeutic targets to protect cerebrovascular endothelium from apoptotic injury following cerebral ischemia.
Collapse
Affiliation(s)
- Yunhong Zhang
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | |
Collapse
|
44
|
Matsumoto T, O'Malley K, Efron PA, Burger C, McAuliffe PF, Scumpia PO, Uchida T, Tschoeke SK, Fujita S, Moldawer LL, Hemming AW, Foley DP. Interleukin-6 and STAT3 protect the liver from hepatic ischemia and reperfusion injury during ischemic preconditioning. Surgery 2006; 140:793-802. [PMID: 17084723 DOI: 10.1016/j.surg.2006.04.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2006] [Revised: 04/27/2006] [Accepted: 04/28/2006] [Indexed: 12/26/2022]
Abstract
BACKGROUND Ischemic preconditioning has been shown to protect the liver from ischemia/reperfusion injury. We hypothesized that IL-6 directly modulates the protective effects of ischemic preconditioning. METHODS Three weeks after undergoing splenic transposition, wild-type C57BL/6 and IL-6 null mice underwent 75 minutes of total hepatic ischemia with or without prior ischemic preconditioning (10 minutes of ischemia followed by 15 minutes of reperfusion). After reperfusion, serum ALT, serum IL-6, hepatic IL-6 mRNA, hepatic pSTAT3, and liver histology were evaluated. RESULTS In wild-type mice, survival at 24 hours was greater in the preconditioned group compared with the non-preconditioned group (75% vs 40%, P<.05). In IL-6 null mice, however, ischemic preconditioning did not improve survival when compared with the non-preconditioned group. Preconditioning significantly reduced hepatocellular injury in wild-type mice (P<.05) when compared with IL-6 null animals. This protection was associated with significant increases in serum IL-6, hepatic IL-6 mRNA, and hepatic pSTAT3 levels (P<.05). The protective effects of ischemic preconditioning that correlated with significant increases in systemic IL-6, hepatic IL-6 mRNA abundance, and pSTAT3 levels, were not observed in IL-6 null mice. CONCLUSIONS The protective effects of ischemic preconditioning during total hepatic ischemia/reperfusion injury are dependent on IL-6 signaling and are associated with increased phosphorylation of hepatic STAT3.
Collapse
Affiliation(s)
- Tadashi Matsumoto
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA, and Department of Surgery, Showa University Fujigaoka Hospital, Yokohama, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Hillion JA, Li Y, Maric D, Takanohashi A, Klimanis D, Barker JL, Hallenbeck JM. Involvement of Akt in preconditioning-induced tolerance to ischemia in PC12 cells. J Cereb Blood Flow Metab 2006; 26:1323-31. [PMID: 16511503 PMCID: PMC1855183 DOI: 10.1038/sj.jcbfm.9600286] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The serine-threonine protein kinase Akt has been identified as an important mediator of cell survival able to counteract apoptotic stimuli. However, hibernation, a model of natural tolerance to cerebral ischemia, is associated with downregulation of Akt. We previously established a model of ischemic tolerance in a PC12 cell line and using this model we now addressed the question whether ischemic tolerance also downregulates Akt in PC12 cells. Kinetic studies showed decreased Akt phosphorylation in tolerized cells. Similarly, phosphorylated levels of three major targets of Akt and well-known proapoptotic factors, the glycogen synthase kinase 3 (GSK-3), a Forkhead family member, FoxO4, and the protein murine double minute 2 (MDM2), all inactivated upon phosphorylation by Akt, were decreased in preconditioned cells. In addition, pharmacological blockade of the phosphoinositide 3-kinase (PI3K)/Akt pathway reduced cell death induced by oxygen and glucose deprivation (OGD) and increased the protective effect of preconditioning (PC). Furthermore, decreasing availability of P-Akt by transfecting PC12 cells with constructs of inactive Akt also resulted in protection against OGD and potentiation of the protective effect of PC. Depending on the environment, GSK-3, FOXO-4, and MDM2 can trigger apoptotic responses or cell cycle arrest, and thus, in a situation of reduced energy, driving the cells into a state of quiescence might be neuroprotective. This work suggests that in the context of tolerance downregulation of Akt is beneficial.
Collapse
Affiliation(s)
- Joëlle A Hillion
- 1Stroke Branch, Department of Health and Human Services, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892-4476, USA.
| | | | | | | | | | | | | |
Collapse
|
46
|
Izuishi K, Tsung A, Hossain MA, Fujiwara M, Wakabayashi H, Masaki T, Billiar TR, Maeta H. Ischemic preconditioning of the murine liver protects through the Akt kinase pathway. Hepatology 2006; 44:573-80. [PMID: 16941697 DOI: 10.1002/hep.21298] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Hepatic ischemia-reperfusion (I/R) injury occurs in the settings of transplantation, trauma, and elective liver resection. Ischemic preconditioning has been used as a strategy to reduce inflammation and organ damage from I/R of the liver. However, the mechanisms involved in this process are poorly understood. We examined the role of the phosphatidylinositol 3 (PI3) kinase/Akt-signaling pathway during hepatic ischemic preconditioning (IPC). Prior to a prolonged warm ischemic insult, BALB/c mice were subjected to a 20-minute IPC period consisting of 10 minutes of ischemia and 10 minutes of reperfusion. Mice undergoing IPC demonstrated a significantly greater level and earlier activation of Akt in the liver compared with control animals. IPC also resulted in markedly less hepatocellular injury and improved survival compared with control animals. Akt activation associated with hepatic IPC suppressed the activity of several modulators of apoptosis, including Bad, glycogen synthase kinase beta, and caspase-3. In addition, IPC also inhibited the activities of c-Jun N-terminal kinase and nuclear factor kappaB after I/R. Pretreatment of mice with PI3 kinase inhibitors completely abolished Akt phosphorylation and the protective effects seen with IPC. In conclusion, these results indicate that the PI3 kinase/Akt pathway plays an essential role in the protective effects of IPC in hepatic I/R injury. Modulation of this pathway may be a potential strategy in clinical settings of ischemic liver injury to decrease organ damage.
Collapse
Affiliation(s)
- Kunihiko Izuishi
- First Department of Surgery, Faculty of Medicine, Kagawa University, Kagawa, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Carini R, Alchera E, De Cesaris MG, Splendore R, Piranda D, Baldanzi G, Albano E. Purinergic P2Y2 receptors promote hepatocyte resistance to hypoxia. J Hepatol 2006; 45:236-45. [PMID: 16644060 DOI: 10.1016/j.jhep.2006.02.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2005] [Revised: 02/10/2006] [Accepted: 02/21/2006] [Indexed: 12/04/2022]
Abstract
BACKGROUND/AIMS ATP stimulation of purinergic P2 receptors (P2YR and P2XR) regulates several hepatic functions. Here we report the involvement of ATP-mediated signals in enhancing hepatocyte tolerance to lethal stress. METHODS The protection given by purinergic agonists was investigated in rat hepatocytes exposed to hypoxia. RESULTS ATP released after hypotonic stress (200 mOsm/L) as well as P2YR agonists prevented hepatocyte killing by hypoxia with efficiency ranking UTP > ATPgammaS > ADPbetaS, whereas the P2XR agonist, methylene-adenosine-5'-triphosphate, was ineffective. Adenosine-5'-O-3-thiotriphosphate (ATPgammaS; 100 micromol/L) also prevented Na+ -overload in hypoxic cells by inhibiting the Na+/H+ exchanger, without interfering with hypoxic acidosis. ATPgammaS activated Src and promoted a Src-dependent stimulation of both ERK1/2 and p38MAPK. Blocking p38MAPK with SB203580 reverted the protection given by ATPgammaS on both cell viability and Na+ accumulation, whereas ERK1/2 inhibition with PD98058 was ineffective. An increased phosphorylation of ERK1/2 was also evident in untreated hypoxic hepatocytes. PD98058 ameliorated Na+ accumulation and cell death caused by hypoxia. Hepatocyte pre-treatment with ATPgammaS reverted ERK1/2 activation in hypoxic cells. SB203580 blocked the effects of ATPgammaS on both ERK1/2 and Na+/H+ exchanger. CONCLUSIONS The activation of p38MAPK by P2Y2R increases hepatocyte resistance to hypoxia by down-modulating ERK1/2-mediated signals that promote Na+ influx through the Na+/H+ exchanger.
Collapse
Affiliation(s)
- Rita Carini
- Department of Medical Sciences, University A. Avogadro of East Piedmont, Novara, Italy.
| | | | | | | | | | | | | |
Collapse
|
48
|
López-Neblina F, Toledo-Pereyra LH. Phosphoregulation of Signal Transduction Pathways in Ischemia and Reperfusion. J Surg Res 2006; 134:292-9. [PMID: 16519903 DOI: 10.1016/j.jss.2006.01.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2005] [Revised: 12/23/2005] [Accepted: 01/06/2006] [Indexed: 01/08/2023]
Abstract
Ischemia/reperfusion (I/R) injury triggered by pathogenic processes, such as organ transplant dysfunction, stroke, myocardial infarction, and shock, stimulate both immune and inflammatory pathways. Inflammatory cell activation and cytotoxic cytokine expression are associated with reperfusion injury. The activation of these inflammatory mediators initiates several interconnected downstream cascades regulated by phosphorylation and dephosphorylation reactions. These complex phosphorylation-dependent signal transduction pathways ultimately initiate nuclear transcription of inflammatory as well as anti-inflammatory genes to repair and assist in the recovery of damaged cells. Radical oxygen species (ROS) production, under ischemic conditions, initiates a cascade of events regulated by phosphorylation/dephosphorylation reactions and inflammatory gene expression. This is a review of the current understanding of the phosphoregulatory mechanisms that mediate the complex processes of signal transduction secondary to I/R injury. The rationale for inhibiting or activating signaling pathways as a promising molecular target for ameliorating reperfusion injury in I/R-related diseases, such as stroke, myocardial infarction, and storage for transplantation, is discussed on the basis of a new understanding of the mechanisms modulating phosphoregulatory pathways. In addition, we present part of our ongoing research in this field with phosphoregulatory signal transduction and its potential application.
Collapse
Affiliation(s)
- Fernando López-Neblina
- Trauma, Surgery Research and Molecular Biology, Borgess Research Institute, Kalamazoo, MI 49048, USA
| | | |
Collapse
|
49
|
Carini R, Trincheri NF, Alchera E, De Cesaris MG, Castino R, Splendore R, Albano E, Isidoro C. PI3K-dependent lysosome exocytosis in nitric oxide-preconditioned hepatocytes. Free Radic Biol Med 2006; 40:1738-48. [PMID: 16678013 DOI: 10.1016/j.freeradbiomed.2006.01.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2005] [Revised: 12/23/2005] [Accepted: 01/09/2006] [Indexed: 11/19/2022]
Abstract
We investigated the signal mediators and the cellular events involved in the nitric oxide (NO)-induced hepatocyte resistance to oxygen deprivation in isolated hepatocytes treated with the NO donor (Z)-1-(N-methyl-N-[6-(N-methylammoniohexyl)amino])diazen-1-ium-1,2-diolate (NOC-9). NOC-9 greatly induced PI3K activation, as tested by phosphorylation of PKB/Akt. This effect was prevented by either 1H-(1,2,4)-oxadiazolo-(4,3)-quinoxalin-1-one, an inhibitor of the soluble guanylate cyclase (sGC), or KT5823, an inhibitor of cGMP-dependent kinase (cGK), as well as by farnesyl protein transferase inhibitor, which blocks the function of Ras GTPase. Bafilomycin A, an inhibitor of the lysosome-type vacuolar H+-ATPase, cytochalasin D, which disrupts the cytoskeleton-dependent organelle traffic, and wortmannin, which inhibits the PI3K-dependent traffic of lysosomes, all abolished the NOC-9-induced hepatocyte protection. The treatment with NOC-9 was associated with the PI3K-dependent peripheral translocation and fusion with the plasma membrane of lysosomes and the appearance at the cell surface of the vacuolar H+-ATPase. Inhibition of sGC, cGK, and Ras, as well as the inhibition of PI3K by wortmannin, prevented the exocytosis of lysosomes and concomitantly abolished the protective effect of NOC-9 on hypoxia-induced pHi and [Na+]i alterations and cell death. These data indicate that NO increases hepatocyte resistance to hypoxic injury by activating a pathway involving Ras, sGC, and cGK that determines PI3K-dependent exocytosis of lysosomes.
Collapse
Affiliation(s)
- Rita Carini
- Laboratory of Pathology, Dipartimento di Scienze Mediche, Università del Piemonte Orientale A. Avogadro, Via Solaroli 17, 28100 Novara, Italy
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Pérez LM, Milkiewicz P, Elias E, Coleman R, Sánchez Pozzi EJ, Roma MG. Oxidative Stress Induces Internalization of the Bile Salt Export Pump, Bsep, and Bile Salt Secretory Failure in Isolated Rat Hepatocyte Couplets: A Role for Protein Kinase C and Prevention by Protein Kinase A. Toxicol Sci 2006; 91:150-8. [PMID: 16452108 DOI: 10.1093/toxsci/kfj113] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We have shown that Ca2+-mediated protein kinase C (PKC) activation induces impairment of bile salt secretory function and F-actin redistribution in hepatocyte couplets. Because oxidative stress induces Ca2+ elevation, we tested here whether PKC inhibition or protein kinase A (PKA) activation, which often counteracts PKC-dependent effects, can prevent and reverse these alterations. The pro-oxidant compounds tert-butylhydroperoxide (tBOOH, 100 microM) and 2,3-dimethoxy-1,4-naphthoquinone (30 microM), reduced by -41% and -29%, respectively, the percentage of couplets accumulating the fluorescent bile salt analog, cholyl-lysylfluorescein in their canalicular vacuoles (p < 0.01). tBOOH-induced bile salt secretory failure was accompanied by internalization of the canalicular bile salt export pump (Bsep), and disarrangement of cytoskeletal F-actin. All these deleterious effects were fully prevented by the intracellular Ca2+ chelator BAPTA/AM (20 microM), the pan-specific PKC inhibitors H7 (100 microM) and staurosporine (1 microM), the inhibitor of Ca2+-dependent PKCs, Gö6976 (2 microM), and the PKA activator dibutyryl-cAMP (500 microM). H7, Gö6976, and dibutyryl-cAMP not only prevented but also fully reversed the decrease in the cholyl-lysyl-fluorescein accumulation. In conclusion, these results suggest that low levels of oxidative stress impair bile salt secretion by internalizing Bsep through a Ca2+-dependent, PKC-mediated mechanism, and that inhibition of PKC, or activation of PKA, prevents and reverses these effects. Alterations in actin organization may be a causal factor.
Collapse
Affiliation(s)
- Leonardo M Pérez
- Institute of Experimental Physiology, CONICET-National University of Rosario, Rosario, Argentina
| | | | | | | | | | | |
Collapse
|