Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Int J Mol Sci ; 22(13)2021 Jun 22.
Article in English | MEDLINE | ID: mdl-34206365

ABSTRACT

Acute liver failure (ALF) is associated with deregulated nitric oxide (NO) signaling in the brain, which is one of the key molecular abnormalities leading to the neuropsychiatric disorder called hepatic encephalopathy (HE). This study focuses on the effect of ALF on the relatively unexplored endothelial NOS isoform (eNOS). The cerebral prefrontal cortices of rats with thioacetamide (TAA)-induced ALF showed decreased eNOS expression, which resulted in an overall reduction of NOS activity. ALF also decreased the content of the NOS cofactor, tetrahydro-L-biopterin (BH4), and evoked eNOS uncoupling (reduction of the eNOS dimer/monomer ratio). The addition of the NO precursor L-arginine in the absence of BH4 potentiated ROS accumulation, whereas nonspecific NOS inhibitor L-NAME or EDTA attenuated ROS increase. The ALF-induced decrease of eNOS content and its uncoupling concurred with, and was likely causally related to, both increased brain content of reactive oxidative species (ROS) and decreased cerebral cortical blood flow (CBF) in the same model.


Subject(s)
Biopterins/analogs & derivatives , Cerebral Cortex/enzymology , Hepatic Encephalopathy/enzymology , Liver Failure, Acute/enzymology , Nitric Oxide Synthase Type III/genetics , Animals , Arginine/metabolism , Biopterins/analysis , Biopterins/metabolism , Cerebral Cortex/blood supply , Cerebral Cortex/metabolism , Cerebrovascular Circulation , Gene Expression Regulation , Hepatic Encephalopathy/etiology , Hepatic Encephalopathy/genetics , Liver Failure, Acute/chemically induced , Liver Failure, Acute/complications , Liver Failure, Acute/genetics , Male , Rats , Rats, Sprague-Dawley , Thioacetamide/toxicity
2.
J Pathol ; 247(1): 110-122, 2019 01.
Article in English | MEDLINE | ID: mdl-30264435

ABSTRACT

Ibuprofen is a worldwide used non-steroidal anti-inflammatory drug which may cause acute liver injury (ALI) requiring liver transplantation. We aimed to unveil the molecular pathways involved in triggering ibuprofen-induced ALI, which, at present, remain elusive. First, we investigated activation of essential pathways in human liver sections of ibuprofen-induced ALI. Next, we assessed the cytotoxicity of ibuprofen in vitro and developed a novel murine model of ibuprofen intoxication. To assess the role of JNK, we used animals carrying constitutive deletion of c-Jun N-terminal kinase 1 (Jnk1-/- ) or Jnk2 (Jnk2-/- ) expression and included investigations using animals with hepatocyte-specific Jnk deletion either genetically (Jnk1Δhepa ) or by siRNA (siJnk2Δhepa ). We found in human and murine samples of ibuprofen-induced acute liver failure that JNK phosphorylation was increased in the cytoplasm of hepatocytes and other non-liver parenchymal cells (non-LPCs) compared with healthy tissue. In mice, ibuprofen intoxication resulted in a significantly stronger degree of liver injury compared with vehicle-treated controls as evidenced by serum transaminases, and hepatic histopathology. Next, we investigated molecular pathways. PKCα, AKT, JNK and RIPK1 were significantly increased 8 h after ibuprofen intoxication. Constitutive Jnk1-/- and Jnk2-/- deficient mice exhibited increased liver dysfunction compared to wild-type (WT) animals. Furthermore, siJnk2Δhepa animals showed a dramatic increase in biochemical markers of liver function, which correlated with significantly higher serum liver enzymes and worsened liver histology, and MAPK activation compared to Jnk1Δhepa or WT animals. In our study, cytoplasmic JNK activation in hepatocytes and other non-LPCs is a hallmark of human and murine ibuprofen-induced ALI. Functional in vivo analysis demonstrated a protective role of hepatocyte-specific Jnk2 during ibuprofen ALI. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Chemical and Drug Induced Liver Injury/prevention & control , Hepatocytes/enzymology , Ibuprofen , Liver Failure, Acute/prevention & control , Liver/enzymology , Mitogen-Activated Protein Kinase 9/metabolism , Animals , Cell Death , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Enzyme Activation , Hepatocytes/pathology , Humans , Liver/pathology , Liver Failure, Acute/enzymology , Liver Failure, Acute/genetics , Liver Failure, Acute/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/genetics , Phosphorylation , Signal Transduction
3.
Apoptosis ; 24(11-12): 934-945, 2019 12.
Article in English | MEDLINE | ID: mdl-31576482

ABSTRACT

Acetaminophen (APAP)-induced acute liver failure (ALF) is a life-threatening disease with only a few treatment options available. Though extensive research has been conducted for more than 40 years, the underlying pathomechanisms are not completely understood. Here, we studied as to whether APAP-induced ALF can be prevented in mice by silencing the BH3-interacting domain death agonist (Bid) as a potential key player in APAP pathology. For silencing Bid expression in mice, siRNABid was formulated with the liver-specific siRNA delivery system DBTC and administered 48 h prior to APAP exposure. Mice which were pre-treated with HEPES (vehicleHEPES) and siRNALuci served as siRNA controls. Hepatic pathology was assessed by in vivo fluorescence microscopy, molecular biology, histology and laboratory analysis 6 h after APAP or PBS exposure. Application of siRNABid caused a significant decrease of mRNA and protein expression of Bid in APAP-exposed mice. Off-targets, such as cytochrome P450 2E1 and glutathione, which are known to be consumed under APAP intoxication, were comparably reduced in all APAP-exposed mice, underlining the specificity of Bid silencing. In APAP-exposed mice non-sterile inflammation with leukocyte infiltration and perfusion failure remained almost unaffected by Bid silencing. However, the Bid silencing reduced hepatocellular damage, evident by a remarkable decrease of DNA fragmented cells in APAP-exposed mice. In these mice, the expression of the pro-apoptotic protein Bax, which recently gained importance in the cell death pathway of regulated necrosis, was also significantly reduced, in line with a decrease in both, necrotic liver tissue and plasma transaminase activities. In addition, plasma levels of HMGB1, a marker of sterile inflammation, were significantly diminished. In conclusion, the liver-specific silencing of Bid expression did not protect APAP-exposed mice from microcirculatory dysfunction, but markedly protected the liver from necrotic cell death and in consequence from sterile inflammation. The study contributes to the understanding of the molecular mechanism of the APAP-induced pathogenic pathway by strengthening the importance of Bid and Bid silencing associated effects.


Subject(s)
Acetaminophen/toxicity , Apoptosis/genetics , BH3 Interacting Domain Death Agonist Protein/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Hepatocytes/metabolism , Liver Failure, Acute/chemically induced , Animals , BH3 Interacting Domain Death Agonist Protein/genetics , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/genetics , Cytochrome P450 Family 2/metabolism , Glutathione/metabolism , HMGB1 Protein/metabolism , Hepatocytes/pathology , Inflammation/complications , Inflammation/metabolism , Liver/enzymology , Liver/metabolism , Liver/pathology , Liver Failure, Acute/enzymology , Liver Failure, Acute/metabolism , Male , Mice , Mice, Inbred C57BL , Necrosis/metabolism , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , bcl-2-Associated X Protein/metabolism
4.
Biochem Biophys Res Commun ; 508(3): 682-689, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30528392

ABSTRACT

In recent years, transplantation of mesenchymal stem cells (MSCs) has attracted much attention as a potential cell-based therapy for acute liver failure (ALF). As an inducible enzyme, heme oxygenase 1 (HO-1) has been reported to have cytoprotective, anti-apoptotic and immunoregulatory effects. Autophagy, a conserved catabolic process in cells, may be an important pathway for MSCs to treat ALF. In this study, we aimed to explore whether MSCs treat ALF by regulating autophagy and whether HO-1 was involved in the same pathway. Bone marrow-derived MSCs were isolated from Sprague-Dawley rats and cultured according to an established protocol. Co-culture systems of MSCs and hepatocytes were used to assess autophagy in the treatment of ALF. Meanwhile, MSCs were transplanted into rats with d-galactosamine (Gal)-induced ALF. Autophagy inhibitor (3-methyladenine, 3-MA), HO-1 inhibitor (zinc protoporphyrin, ZnPP) and PI3K specific inhibitor (LY294002) were employed in the study. Blood samples and liver tissues were collected before euthanasia. Survival rate, liver function, inflammatory factors, histology, Ki67 and TUNEL staining were determined. MSCs transplantation alleviated ALF both in vivo and in vitro. Autophagy and autophagy-related proteins were significantly up-regulated during MSCs treatment. 3-MA attenuated the therapeutic effect of MSCs. Administration of LY294002 before ALF induction inhibited hepatocyte autophagy. During the MSCs treatment, the HO-1 expression was increased, while inhibiting HO-1 attenuated the therapeutic effect of MSCs as well as hepatocyte autophagy. These findings suggested MSCs could alleviate ALF by increasing the HO-1 expression, which played an important role in activating autophagy through PI3K/AKT signaling pathway.


Subject(s)
Autophagy , Heme Oxygenase-1/metabolism , Liver Failure, Acute/enzymology , Liver Failure, Acute/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Animals , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Hepatocytes/pathology , Hepatocytes/ultrastructure , Inflammation/pathology , Liver/injuries , Liver/pathology , Liver Failure, Acute/pathology , Male , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Signal Transduction , Up-Regulation
5.
Am J Physiol Renal Physiol ; 314(5): F942-F955, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29357415

ABSTRACT

We have reported that tumor necrosis factor-α (TNF-α) is critical for reduction of glomerular filtration rate (GFR) in rats with fulminant hepatic failure (FHF). The present study aims to evaluate the underlying mechanisms of decreased GFR during acute hepatic failure. Rats with FHF induced by d-galactosamine plus lipopolysaccharide (GalN/LPS) were injected intravenously with recombinant lentivirus harboring short hairpin RNA against the protein kinase C-α ( PKC-α) gene (Lenti-shRNA-PKC-α). GFR, serum levels of aminotransferases, creatinine, urea nitrogen, potassium, sodium, chloride, TNF-α, and endothelin-1 (ET-1), as well as type 1 inositol 1,4,5-trisphosphate receptor (IP3R1) expression in renal tissue were assessed. The effects of PKC-α silencing on TNF-α-induced IP3R1, specificity protein 1 (SP-1), and c-Jun NH2-terminal kinase (JNK) expression, as well as cytosolic calcium content were determined in glomerular mesangial cell (GMCs) with RNAi against PKC-α. Renal IP3R1 overexpression was abrogated by pre-treatment with Lenti-shRNA-PKC-α. The PKC-α silence significantly improved the compromised GFR, reduced Cr levels, and reversed the decrease in glomerular inulin space and the increase in glomerular calcium content in GalN/LPS-exposed rats. TNF-α treatment increased expression of PKC-α, IP3R1, specificity protein 1 (SP-1), JNK, and p-JNK in GMCs and increased Ca2 + release and binding activity of SP-1 to the IP3R1 promoter. These effects were blocked by transfection of siRNA against the PKC-α gene, and the PKC-α gene silence also restored cytosolic Ca2+ concentration. RNAi targeting PKC-α inhibited TNF-α-induced IP3R1 overexpression and in turn improved compromised GFR in the development of acute kidney injury during FHF in rats.


Subject(s)
Acute Kidney Injury/therapy , Chemical and Drug Induced Liver Injury/therapy , Glomerular Filtration Rate/drug effects , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Kidney/drug effects , Liver Failure, Acute/therapy , Protein Kinase C-alpha/metabolism , RNA Interference , RNA, Small Interfering/administration & dosage , Tumor Necrosis Factor-alpha/pharmacology , Acute Kidney Injury/enzymology , Acute Kidney Injury/physiopathology , Animals , Cell Line , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/physiopathology , Disease Models, Animal , Inositol 1,4,5-Trisphosphate Receptors/genetics , Kidney/enzymology , Kidney/physiopathology , Liver Failure, Acute/enzymology , Liver Failure, Acute/physiopathology , Male , Protein Kinase C-alpha/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects
6.
BMC Anesthesiol ; 18(1): 71, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29925334

ABSTRACT

BACKGROUND: Acute liver failure (ALF) is a life-threatening entity particularly when infectious complications worsen the clinical course. Urgent liver transplantation (LT) is frequently the only curative treatment. However, in some cases, recovery is observed under conservative treatment. Therefore, prognostic tools for estimating course of the disease are of great clinical interest. Since laboratory parameters sometimes lack sensitivity and specificity, enzymatic liver function measured by liver maximum capacity (LiMAx) test may offer novel and valuable additional information in this setting. CASE PRESENTATION: We here report the case of a formerly healthy 20-year old male caucasian patient who was admitted to our clinic for ALF of unknown origin in December 2017. Laboratory parameters confirmed the diagnosis with an initial MELD score of 28 points. Likewise, enzymatic liver function was significantly impaired with a value of 147 [> 315] µg/h/kg. Clinical and biochemical analyses for viral-, autoimmune-, or drug-induced hepatitis were negative. Liver synthesis parameters further deteriorated reaching a MELD score of 40 points whilst clinical course was complicated by septic pneumonia leading to severe hepatic encephalopathy grade III-IV, finally resulting in mechanical ventilation of the patient. Interestingly, although clinical course and laboratory data suggested poor outcome, serial LiMAx test revealed improvement of the enzymatic liver function at this time point increasing to 169 µg/h/kg. Clinical condition and laboratory data slowly improved likewise, however with significant time delay of 11 days. Finally, the patient could be dismissed from our clinic after 37 days. CONCLUSION: Estimating prognosis in patients with ALF is challenging by use of the established scores. In our case, improvement of enzymatic liver function measured by the LiMAx test was the first parameter predicting beneficial outcome in a patient with ALF complicated by sepsis.


Subject(s)
Liver Failure, Acute/diagnosis , Liver Function Tests/statistics & numerical data , Predictive Value of Tests , Humans , Liver Failure, Acute/complications , Liver Failure, Acute/enzymology , Male , Prognosis , Sepsis/complications , Time Factors , Young Adult
7.
Gastroenterology ; 150(4): 968-81, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26708719

ABSTRACT

BACKGROUND & AIMS: c-Jun N-terminal kinase (JNK) 1 and JNK2 are expressed in hepatocytes and have overlapping and distinct functions. JNK proteins are activated via phosphorylation in response to acetaminophen- or carbon tetrachloride (CCl4)-induced liver damage; the level of activation correlates with the degree of injury. SP600125, a JNK inhibitor, has been reported to block acetaminophen-induced liver injury. We investigated the role of JNK in drug-induced liver injury (DILI) in liver tissue from patients and in mice with genetic deletion of JNK in hepatocytes. METHODS: We studied liver sections from patients with DILI (due to acetaminophen, phenprocoumon, nonsteroidal anti-inflammatory drugs, or autoimmune hepatitis) or patients without acute liver failure (controls) collected from a DILI Biobank in Germany. Levels of total and activated (phosphorylated) JNK were measured by immunohistochemistry and Western blotting. Mice with hepatocyte-specific deletion of Jnk1 (Jnk1(Δhepa)) or combination of Jnk1 and Jnk2 (Jnk(Δhepa)), as well as Jnk1-floxed C57BL/6 (control) mice, were given injections of CCl4 (to induce fibrosis) or acetaminophen (to induce toxic liver injury). We performed gene expression microarray and phosphoproteomic analyses to determine mechanisms of JNK activity in hepatocytes. RESULTS: Liver samples from DILI patients contained more activated JNK, predominantly in nuclei of hepatocytes and in immune cells, than healthy tissue. Administration of acetaminophen to Jnk(Δhepa) mice produced a greater level of liver injury than that observed in Jnk1(Δhepa) or control mice, based on levels of serum markers and microscopic and histologic analysis of liver tissues. Administration of CCl4 also induced stronger hepatic injury in Jnk(Δhepa) mice, based on increased inflammation, cell proliferation, and fibrosis progression, compared with Jnk1(Δhepa) or control mice. Hepatocytes from Jnk(Δhepa) mice given acetaminophen had an increased oxidative stress response, leading to decreased activation of adenosine monophosphate-activated protein kinase, total protein adenosine monophosphate-activated protein kinase levels, and pJunD and subsequent necrosis. Administration of SP600125 before or with acetaminophen protected Jnk(Δhepa) and control mice from liver injury. CONCLUSIONS: In hepatocytes, JNK1 and JNK2 appear to have combined effects in protecting mice from CCl4- and acetaminophen-induced liver injury. It is important to study the tissue-specific functions of both proteins, rather than just JNK1, in the onset of toxic liver injury. JNK inhibition with SP600125 shows off-target effects.


Subject(s)
Chemical and Drug Induced Liver Injury/prevention & control , Hepatocytes/enzymology , Liver Failure, Acute/prevention & control , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , AMP-Activated Protein Kinases/metabolism , Acetaminophen , Animals , Carbon Tetrachloride , Case-Control Studies , Cell Death , Cell Proliferation , Cells, Cultured , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Enzyme Activation , Female , Gene Expression Profiling , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , Liver Failure, Acute/chemically induced , Liver Failure, Acute/enzymology , Liver Failure, Acute/genetics , Liver Failure, Acute/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Mitogen-Activated Protein Kinase 8/deficiency , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/antagonists & inhibitors , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/genetics , Oxidative Stress , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Time Factors , Young Adult
8.
Acta Pharmacol Sin ; 38(11): 1554-1565, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28770824

ABSTRACT

HIV infection is often associated with liver failure, which alters the pharmacokinetics of many drugs. In this study we investigated whether acute liver failure (ALF) altered the pharmacokinetics of the first-line anti-HIV agent zidovudine (AZT), a P-gp/BCRP substrate, in rats. ALF was induced in rats by injecting thioacetamide (TAA, 300 mg·kg-1·d-1, ip) for 2 days. On the second day after the last injection of TAA, the pharmacokinetics of AZT was investigated following both oral (20 mg/kg) and intravenous (10 mg/kg) administration. ALF significantly increased the plasma concentrations of AZT after both oral and intravenous doses of AZT, but without affecting the urinary excretion of AZT. AZT metabolism was studied in rat hepatic microsomes in vitro, which revealed that hepatic UGT2B7 was the main enzyme responsible for the formation of AZT O-glucuronide (GAZT); ALF markedly impaired AZT metabolism in hepatic microsomes, which was associated with the significantly decreased hepatic UGT2B7 expression. Intestinal absorption of AZT was further studied in rats via in situ single-pass intestinal perfusion. Intestinal P-gp function and intestinal integrity were assessed with rhodamine 123 and FD-70, respectively. We found that ALF significantly downregulated intestinal P-gp expression, and had a smaller effect on intestinal BCRP. Further studies showed that ALF significantly increased the intestinal absorption of both rhodamine 123 and AZT without altering intestinal integrity, thus confirming an impairment of intestinal P-gp function. In conclusion, ALF significantly increases the oral plasma exposure of AZT in rats, a result partly attributed to the impaired function and expression of hepatic UGT2B7 and intestinal P-gp.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Glucuronosyltransferase/metabolism , Jejunum/metabolism , Liver Failure, Acute/enzymology , Liver/enzymology , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/pharmacokinetics , Zidovudine/administration & dosage , Zidovudine/pharmacokinetics , Administration, Intravenous , Administration, Oral , Animals , Biological Availability , Disease Models, Animal , Intestinal Absorption , Male , Microsomes, Liver/enzymology , Rats, Sprague-Dawley , Renal Elimination , Reverse Transcriptase Inhibitors/blood , Thioacetamide , Zidovudine/blood
9.
J Surg Res ; 205(1): 1-10, 2016 09.
Article in English | MEDLINE | ID: mdl-27620992

ABSTRACT

BACKGROUND: Histone deacetylase (HDAC) inhibitors have been widely applied in the clinic as anticancer drugs against multiple neoplasms and proved their anti-inflammation under different pathology recently. Trichostatin A (TSA) is an HDAC inhibitor specific in class I and II HDAC enzymes. The aim of the present study was to elucidate the protective effects of TSA on acute liver failure (ALF) in rats and its potential mechanism. METHODS: A total of 18 female Sprague-Dawley rats were separated into control, model, and TSA groups. We used Western blotting to determine the expression of HDACs, inflammatory cytokines, and acetylation of histone in liver and small intestine. The gene expression of inflammatory factors and Cox-2 was detected by a polymerase chain reaction. Colonic motility was assessed by spatiotemporal mapping. Histologic analysis and immunohistochemistry were performed. Intestinal permeability examination and levels of alanine aminotransferase, aspartate aminotransferase, and total bilirubin were also observed. RESULTS: ALF procedure caused harm to histology of liver and small intestine, increased the intestinal permeability and serum levels of alanine aminotransferase, aspartate aminotransferase, and total bilirubin. It also interrupted the normal organization of colonic motor patterns by hurting enteric nervous system and pacemaker cells. Along with the decrease of inflammatory factors in ALF rats by TSA administration, all the damage to the liver, the small intestine, and the colon was repaired. CONCLUSIONS: TSA alleviates the lesion in liver, as well as in small intestine and colon in ALF rats by directly inhibiting inflammatory response.


Subject(s)
Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Intestinal Diseases/prevention & control , Liver Failure, Acute/complications , Liver/drug effects , Animals , Cytokines/metabolism , Female , Gastrointestinal Motility/drug effects , Histone Deacetylases/metabolism , Intestinal Diseases/etiology , Intestine, Small/drug effects , Lipopolysaccharides , Liver/pathology , Liver Failure, Acute/enzymology , Liver Failure, Acute/pathology , Rats , Rats, Sprague-Dawley
10.
Biochem J ; 466(2): 369-78, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25387528

ABSTRACT

CCAAT/enhancer binding protein (C/EBP)-homologous protein (CHOP) has been shown to be a key molecule in endoplasmic reticulum (ER) stress-mediated apoptosis. ER oxidoreductin 1-α (ERO1α), a target of CHOP, is an important oxidizing enzyme that regulates reactive oxygen species (ROS), which play a prominent role in hepatocellular death during acute liver failure (ALF). However, little is known about how CHOP facilitates ROS-induced hepatocellular injury. The present study was designed to investigate the roles and molecular mechanisms of CHOP in ALF. In the liver tissues from ALF patients, the expression of CHOP was significantly increased, which was accompanied by increased expression of dsRNA-dependent protein kinase (PKR)-like ER kinase (PERK) signalling, activating transcription factor 4 (ATF6) signalling, inositol-requiring enzyme-1 (IRE1) signalling and ERO1α, as compared with healthy controls. In the mouse model of galactosamine (GaIN)/lipopolysaccharide (LPS)-induced ALF, the hepatocellular injury was accompanied by up-regulated PERK signalling, ATF6 signalling, IRE1 signalling, CHOP and ERO1α. In contrast, CHOP deficiency decreased hepatocellular apoptosis/necrosis and increased animal survival. Furthermore, disruption of CHOP decreased ERO1α expression leading to reducing ROS-induced cell death in vivo and in vitro. Interestingly, ERO1α overexpression restored GaIN/LPS-induced hepatocellular injury in CHOP-deficient mice. Our studies demonstrate for the first time that CHOP promotes liver damage during ALF through activation of ERO1α, a key mediator to link ER stress and ROS. Therefore, targeting CHOP/ERO1α signalling could be a novel therapeutic approach during ALF.


Subject(s)
Glycoproteins/metabolism , Hepatocytes/metabolism , Liver Failure, Acute/metabolism , MAP Kinase Signaling System , Membrane Glycoproteins/metabolism , Oxidoreductases/metabolism , Transcription Factor CHOP/metabolism , Adult , Animals , Apoptosis , Cells, Cultured , Disease Models, Animal , Female , Gene Silencing , Glycoproteins/antagonists & inhibitors , Glycoproteins/genetics , Hepatocytes/enzymology , Hepatocytes/pathology , Humans , Liver Failure, Acute/enzymology , Liver Failure, Acute/pathology , Male , Membrane Glycoproteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Necrosis , Oxidoreductases/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Survival Analysis , Transcription Factor CHOP/genetics , Up-Regulation
11.
Immunopharmacol Immunotoxicol ; 38(2): 61-7, 2016.
Article in English | MEDLINE | ID: mdl-26630551

ABSTRACT

Acute hepatic failure involves in excessive oxidative stress and inflammatory responses, leading to a high mortality due to lacking effective therapy. Resolvin D1 (RvD1), an endogenous lipid mediator derived from polyunsaturated fatty acids, has been shown anti-inflammatory and anti-oxidative actions, however, whether RvD1 has protective effects on hepatic failure remains elusive. In this study, the roles and molecular mechanisms of RvD1 were explored in carbon tetrachloride (CCl4)-induced acute liver injury. Our results showed that RvD1 protected mice against CCl4-induced hepatic damage, as evaluated by reduced aminotransferase activities and malondialdehyde content, elevated glutathione and superoxide dismutase activities, and alleviated hepatic pathological damage. Moreover, RvD1 significantly attenuated serum tumor necrosis factor-α and interleukin-6 levels as well as hepatic myeloperoxidase activity, whereas enhanced serum IL-10 level in CCl4-administered mice. Further, RvD1 markedly up-regulated the expression and activity of heme oxygenase-1 (HO-1). However, inhibition of HO-1 activity reversed the protective effects of RvD1 on CCl4-induced liver injury. These results suggest that RvD1 could effectively prevent CCl4-induced liver injury by inhibition of oxidative stress and inflammation, and the underlying mechanism may be related to up-regulation of HO-1.


Subject(s)
Carbon Tetrachloride Poisoning , Docosahexaenoic Acids/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Heme Oxygenase-1/biosynthesis , Liver Failure, Acute , Membrane Proteins/biosynthesis , Up-Regulation/drug effects , Animals , Carbon Tetrachloride Poisoning/enzymology , Carbon Tetrachloride Poisoning/pathology , Carbon Tetrachloride Poisoning/prevention & control , Cytokines/biosynthesis , Liver Failure, Acute/enzymology , Liver Failure, Acute/pathology , Liver Failure, Acute/prevention & control , Male , Mice , Mice, Inbred BALB C , Oxidative Stress/drug effects
12.
Neurochem Res ; 40(2): 293-300, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25161077

ABSTRACT

Glutathione and thioredoxin are complementary antioxidants in the protection of mammalian tissues against oxidative-nitrosative stress (ONS), and ONS is a principal cause of symptoms of hepatic encephalopathy (HE) associated with acute liver failure (ALF). We compared the activities of the thioredoxin system components: thioredoxin (Trx), thioredoxin reductase (TrxR) and the expression of the thioredoxin-interacting protein, and of the key glutathione metabolizing enzyme, glutathione peroxidase (GPx) in the cerebral cortex of rats with ALF induced by thioacetamide (TAA). ALF increased the Trx and TrxR activity without affecting Trip protein expression, but decreased GPx activity in the brains of TAA-treated rats. The total antioxidant capacity (TAC) of the brain was increased by ALF suggesting that upregulation of the thioredoxin may act towards compensating impaired protection by the glutathione system. Intraperitoneal administration of L-histidine (His), an amino acid that was earlier reported to prevent acute liver failure-induced mitochondrial impairment and brain edema, abrogated most of the acute liver failure-induced changes of both antioxidant systems, and significantly increased TAC of both the control and ALF-affected brain. These observations provide further support for the concept of that His has a potential to serve as a therapeutic antioxidant in HE. Most of the enzyme activity changes evoked by His or ALF were not well correlated with alterations in their expression at the mRNA level, suggesting complex translational or posttranslational mechanisms of their modulation, which deserve further investigations.


Subject(s)
Antioxidants/metabolism , Cerebral Cortex/metabolism , Glutathione Peroxidase/metabolism , Histidine/pharmacology , Liver Failure, Acute/metabolism , Thioredoxins/metabolism , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Liver Failure, Acute/enzymology , Male , Rats , Rats, Sprague-Dawley
13.
Ann Hepatol ; 12(4): 556-62, 2013.
Article in English | MEDLINE | ID: mdl-23813133

ABSTRACT

BACKGROUND: Acute liver failure (ALF) is a rare but potentially life-threatening condition and liver transplantation (LTX) remains frequently the only effective therapy. Nevertheless, some patients recover without LTX but the individual indication for or against LTX remains difficult. AIM: To evaluate maximal liver function capacity (LiMAx) for predicting the prognosis of ALF. Material and methods. Clinic data of 12 patients was retrospectively analyzed to compare the different liver function test results with the patients' clinical outcome. Patients were assessed by the LiMAx test, a non-invasive breath test determining cytochrome P450 1A2 capacity using intravenous 13C-methacetin. Statistical analysis compared patients with spontaneous recovery versus non-recovery (LTX or death). RESULTS: Twelve patients (6 male, 6 female; 49 [11-72] years) with viral hepatitis (n = 2), toxic liver injury (n = 3), or cryptogenic liver failure (n = 7) were analyzed. Seven patients fully recovered from ALF and were discharged without LTX. Three patients died and two underwent LTX. The King's College Criteria (KCC) was fulfilled in only one out of five patients without recovery. The LiMAx was 19 ± 19 (16-62) for non-recovery vs. 94 ± 119 (39-378) µg/kg/h for recovery (P = 0.018). In contrast, all biochemical parameters [bilirubin (P = 0.106), creatinine (P = 0.343), AST (P = 0.53), ALT (P = 0.876) and INR (P = 0.876) were statistically indistinct. Also the Model for End-Stage Liver Disease (MELD) score did not show a difference [35 ± 4.3 (29-40) vs. 30 ± 11.5 (6-40); P = 0.27]. CONCLUSIONS: Maximal liver function capacity determined by LiMAx test is severely impaired in patients with ALF. The LiMAx test might be effective in predicting the individual prognosis and the need for LTX in ALF.


Subject(s)
Breath Tests , Liver Failure, Acute/diagnosis , Liver Function Tests/methods , Acetamides/administration & dosage , Adolescent , Adult , Aged , Area Under Curve , Biomarkers/blood , Child , Cytochrome P-450 CYP1A2/metabolism , Female , Humans , International Normalized Ratio , Liver Failure, Acute/blood , Liver Failure, Acute/enzymology , Liver Failure, Acute/mortality , Liver Failure, Acute/surgery , Liver Transplantation , Male , Middle Aged , Pilot Projects , Predictive Value of Tests , Prognosis , ROC Curve , Recovery of Function , Retrospective Studies , Substrate Specificity , Time Factors , Young Adult
14.
Int J Mol Sci ; 14(6): 11224-37, 2013 May 27.
Article in English | MEDLINE | ID: mdl-23712360

ABSTRACT

Cisplatin is one of the most widely used chemical drugs for anticancer treatment. Recent studies have focused on the ability of cisplatin to retain the high mobility group box 1 (HMGB1) protein in cisplatin-DNA adducts, thereby preventing its release from the nucleus. Because HMGB1 is a powerful inflammatory mediator in many diseases, the aim of this study is to evaluate the therapeutic effect of cisplatin acute liver failure. In this study, low-dose cisplatin was administered to treat PMA-induced macrophage-like cells induced by PMA and rats with acute liver failure. We found that cell viability and liver injury were greatly improved by cisplatin treatment. The extracellular levels of HMGB1, TNF-α and IFN-γ were also significantly decreased by the administration of cisplatin. During inflammation, nuclear HMGB1 translocates from the nucleus to the cytoplasm. The administration of cisplatin reduced the cytoplasmic levels of HMGB1 and increased nuclear HMGB1 levels in vitro and in vivo. In conclusion, cisplatin can protect against acute liver failure by retaining HMGB1 in the nucleus and preventing its release into the extracellular milieu.


Subject(s)
Cell Nucleus/metabolism , Cisplatin/pharmacology , Cisplatin/therapeutic use , Cytoprotection/drug effects , HMGB1 Protein/metabolism , Liver Failure, Acute/drug therapy , Alanine Transaminase/blood , Animals , Apoptosis/drug effects , Cell Line , Cell Nucleus/drug effects , Cell Survival/drug effects , Cytokines/biosynthesis , Liver Failure, Acute/blood , Liver Failure, Acute/enzymology , Liver Failure, Acute/pathology , Macrophages/drug effects , Macrophages/metabolism , Male , Rats, Wistar , Survival Analysis , Time Factors
15.
Am J Hum Genet ; 85(3): 401-7, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19732863

ABSTRACT

Acute liver failure in infancy accompanied by lactic acidemia was previously shown to result from mtDNA depletion. We report on 13 unrelated infants who presented with acute liver failure and lactic acidemia with normal mtDNA content. Four died during the acute episodes, and the survivors never had a recurrence. The longest follow-up period was 14 years. Using homozygosity mapping, we identified mutations in the TRMU gene, which encodes a mitochondria-specific tRNA-modifying enzyme, tRNA 5-methylaminomethyl-2-thiouridylate methyltransferase. Accordingly, the 2-thiouridylation levels of the mitochondrial tRNAs were markedly reduced. Given that sulfur is a TRMU substrate and its availability is limited during the neonatal period, we propose that there is a window of time whereby patients with TRMU mutations are at increased risk of developing liver failure.


Subject(s)
Liver Failure, Acute/enzymology , Liver Failure, Acute/genetics , Mitochondrial Proteins/genetics , Mutation/genetics , tRNA Methyltransferases/genetics , DNA, Mitochondrial/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Genotype , Humans , Infant , Infant, Newborn , Liver/pathology , Liver Failure, Acute/pathology , Mitochondria/enzymology , Protein Biosynthesis , RNA, Transfer/metabolism , Sulfhydryl Compounds/metabolism
16.
Mol Genet Metab ; 106(3): 351-8, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22607940

ABSTRACT

Infantile hepatopathies are life-threatening liver disorders that manifest in the first few months of life. We report on a consanguineous Irish Traveller family that includes six individuals presenting with acute liver failure in the first few months of life. Additional symptoms include anaemia, renal tubulopathy, developmental delay, seizures, failure to thrive and deterioration of liver function with minor illness. The multisystem manifestations suggested a possible mitochondrial basis to the disorder. However, known causes of childhood liver failure and mitochondrial disease were excluded in this family by biochemical, metabolic and genetic analyses. We aimed to identify the underlying risk gene using homozygosity mapping and whole exome sequencing. SNP homozygosity mapping identified a candidate locus at 5q31.3-q33.1. Whole exome sequencing identified 1 novel homozygous missense mutation within the 5q31.3-q33.1 candidate region that segregated with the hepatopathy. The candidate mutation is located in the LARS gene which encodes a cytoplasmic leucyl-tRNA synthetase enzyme responsible for exclusively attaching leucine to its cognate tRNA during protein translation. Knock-down of LARS in HEK293 cells did not impact on mitochondrial function even when the cells were put under physiological stress. The molecular studies confirm the findings of the patients' biochemical and genetic analyses which show that the hepatopathy is not a mitochondrial-based dysfunction problem, despite clinical appearances. This study highlights the clinical utility of homozygosity mapping and exome sequencing in diagnosing recessive liver disorders. It reports mutation of a cytoplasmic aminoacyl-tRNA synthetase enzyme as a possible novel cause of infantile hepatopathy and underscores the need to consider mutations in LARS in patients with liver disease and multisystem presentations.


Subject(s)
Amino Acyl-tRNA Synthetases/genetics , Liver Failure/enzymology , Liver Failure/genetics , Mutation , Adolescent , Adult , Base Sequence , Child , Child, Preschool , Cytoplasm/enzymology , HEK293 Cells , Homozygote , Humans , Infant , Leucine/genetics , Leucine/metabolism , Liver Failure, Acute/enzymology , Liver Failure, Acute/genetics , Mitochondria/metabolism , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/genetics , Molecular Sequence Data , Pedigree , Young Adult
17.
Hepatobiliary Pancreat Dis Int ; 11(4): 383-92, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22893465

ABSTRACT

BACKGROUND: Prior experimentation has shown that loss of the tyrosine kinase (TK) signaling domain of the Ron receptor leads to marked hepatocyte protection in a model of lipopolysaccharide-induced acute liver failure (ALF) in D-galactosamine (GalN)-sensitized mice. The aim of this study was to identify the role of Ron in the regulation of hepatic gene expression. METHODS: Microarray analyses were performed on liver RNA isolated sequentially from wild-type (WT) and TK-/- mice during the progression of ALF. Gene array data were validated using Western and immunohistochemistry analyses as well as with ex vivo culture systems. RESULTS: At baseline, 101 genes were differentially expressed between WT and TK-/- livers, which regulate processes involved in hypoxia, proliferation, apoptosis and metabolism. One hour after ALF induction, WT livers exhibited increased cytokine expression compared to TK-/- livers, and after 4 hours, an induction of suppressor of cytokine signaling (SOCS) genes as well as JAK-STAT pathway activation were prominent in TK-/- livers compared to controls. CONCLUSION: Our studies suggest a novel hepato-protective mechanism in Ron TK-/- mice wherein increased and sustained SOCS production and JAK-STAT activation in the hepatocyte may inhibit the destructive proinflammatory milieu and promote survival factors which blunt hepatic death and the ensuing development of ALF.


Subject(s)
Lipopolysaccharides , Liver Failure, Acute/enzymology , Liver/enzymology , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Apoptosis , Blotting, Western , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Galactosamine , Gene Expression Profiling/methods , Gene Expression Regulation , Immunohistochemistry , Inflammation Mediators/metabolism , Janus Kinases/genetics , Janus Kinases/metabolism , Liver/pathology , Liver Failure, Acute/chemically induced , Liver Failure, Acute/genetics , Liver Failure, Acute/pathology , Liver Failure, Acute/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Receptor Protein-Tyrosine Kinases/deficiency , Receptor Protein-Tyrosine Kinases/genetics , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Time Factors
18.
Am J Pathol ; 176(3): 1400-8, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20075201

ABSTRACT

Brain edema and the associated increase in intracranial pressure are potentially lethal complications of acute liver failure (ALF). Astrocyte swelling (cytotoxic edema) represents a significant component of the brain edema in ALF, and elevated blood and brain ammonia levels have been strongly implicated in its formation. We earlier showed in cultured astrocytes that oxidative stress (OS) and the mitochondrial permeability transition (mPT) play major roles in the mechanism of ammonia-induced astrocyte swelling. Glutamine, a byproduct of ammonia metabolism, has also been shown to induce OS, the mPT, and astrocyte swelling. Such effects of glutamine were suggested to be mediated by its hydrolysis in mitochondria, potentially yielding high levels of ammonia in this organelle and leading to OS and the mPT. L-histidine, an inhibitor of mitochondrial glutamine transport, was recently shown to mitigate OS, mPT, and cell swelling in cultured astrocytes treated with ammonia. The present study examined whether L-histidine similarly abolishes OS, the mPT, and brain edema in a rat model of ALF. Treatment of rats with thioacetamide caused a significant degree of brain edema, which was associated with induction of OS and the mPT. These changes were completely abolished by L-histidine, supporting a key role of mitochondrial glutamine transport and hydrolysis in the mechanism of the brain edema associated with ALF.


Subject(s)
Brain Edema/drug therapy , Brain Edema/etiology , Histidine/therapeutic use , Liver Failure, Acute/complications , Ammonia/metabolism , Animals , Brain/drug effects , Brain/enzymology , Brain/pathology , Brain Edema/enzymology , Glutamate-Ammonia Ligase/metabolism , Glutaminase/metabolism , Glutamine/metabolism , Heme Oxygenase-1/metabolism , Histidine/pharmacology , Liver Failure, Acute/enzymology , Male , Mitochondria/drug effects , Mitochondria/metabolism , Permeability/drug effects , Rats , Rats, Inbred F344
19.
Liver Int ; 31(8): 1209-21, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21745296

ABSTRACT

BACKGROUND: Acute liver failure (ALF) can be induced in mice by administering Escherichia coli lipopolysaccharide (LPS) and D-galactosamine (D-GalN), which induce an inflammatory response involving tumour necrosis factor (TNF)-α production and a hepatocyte-specific transcriptional block. Under these conditions, binding of TNF-α to its cognate receptor on hepatocytes eventually leads to their apoptosis. AIMS: As part of an effort to identify drugs to treat this disease model, we have investigated whether the glutamine synthetase inhibitor methionine sulfoximine (MSO) could play a protective role, given its effectiveness in the inhibition of brain swelling associated with hyperammonaemia. METHODS: Mouse survival, glutamine synthetase activity, hepatocyte apoptosis and induction of inflammatory cytokines were measured in mice treated with MSO before an intraperitoneal injection of LPS/D-GalN. The effect of MSO on viability and on TNF-α release was also assessed on inflammatory and liver cells. RESULTS: We have found that, in mice treated with LPS/D-GalN, MSO (i) drastically increases animal survival; (ii) sharply reduces glutamine synthetase activity, without inhibiting its other target, γ-glutamyl cysteine synthetase; (iii) inhibits death receptor-mediated apoptosis in hepatocytes upstream to cytokine binding; (iv) strongly reduces the overall inflammatory cytokine response, including a significant decrease in TNF-α induction in vivo and ex vivo, and in the interferon-γ level and signalling. CONCLUSIONS: These results demonstrate that the MSO target glutamine synthetase is required for the early steps of the cytokine response to endotoxins, and that its pharmacological inhibition may be exploited to treat inflammation.


Subject(s)
Cytokines/metabolism , Enzyme Inhibitors/pharmacology , Glutamate-Ammonia Ligase/antagonists & inhibitors , Inflammation Mediators/metabolism , Liver Failure, Acute/drug therapy , Liver/drug effects , Methionine Sulfoximine/pharmacology , Animals , Apoptosis/drug effects , Cells, Cultured , Disease Models, Animal , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Female , Galactosamine , Glutamate-Ammonia Ligase/metabolism , Interferon-gamma/metabolism , Lipopolysaccharides , Liver/enzymology , Liver/immunology , Liver/pathology , Liver Failure, Acute/enzymology , Liver Failure, Acute/immunology , Liver Failure, Acute/pathology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Male , Mice , STAT1 Transcription Factor/metabolism , Time Factors , Tumor Necrosis Factor-alpha/metabolism
20.
Liver Int ; 31(8): 1222-30, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21745297

ABSTRACT

BACKGROUND AND AIMS: Acute liver failure (ALF) is frequently complicated by cerebral oedema, systemic inflammation and multiorgan dysfunction. Vascular endothelial growth factor (VEGF) may stimulate liver regeneration but it can also be pro-inflammatory, activating endothelial cells and increasing permeability, actions mediated through Src kinase signalling. We therefore examined whether a Src inhibitor could have therapeutic potential in ALF. METHODS: Murine ALF was induced with azoxymethane. Liver pathology was graded by a blinded examiner and apoptosis quantified by immunohistochemistry. Cerebral VEGF expression was imaged using VEGF-green fluorescent protein transgenic mice. Circulating and macrophage-secreted VEGF levels were measured. Experimental animals received a Src inhibitor or vehicle controls. RESULTS: VEGF was undetectable in normal plasma but reached a mean of 835 pg/ml at grade III encephalopathy (P<0.001). Ammonia, lipopolysaccharide and interferon-gamma acted synergistically to enhance VEGF secretion by macrophages. Production of VEGF by cerebral cortical astrocytes increased with disease progression. Late treatment with inhibitors of Src or VEGF did not improve liver histology, encephalopathy or survival. However, early use of a Src kinase inhibitor significantly reduced hepatic injury, delayed encephalopathy and allowed 25% of mice to survive an otherwise lethal insult. CONCLUSION: Systemic and cerebral VEGF levels are significantly elevated during experimental ALF and may be exacerbated by hyperammonemia and macrophage activation. Early use of a Src inhibitor reduced hepatocellular injury and enabled survival, indicating such agents may have some promise in the treatment of ALF.


Subject(s)
Aniline Compounds/pharmacology , Brain Edema/prevention & control , Brain/drug effects , Liver Failure, Acute/drug therapy , Liver/drug effects , Nitriles/pharmacology , Protein Kinase Inhibitors/pharmacology , Quinolines/pharmacology , Vascular Endothelial Growth Factor A/metabolism , src-Family Kinases/antagonists & inhibitors , Ammonium Chloride/pharmacology , Animals , Apoptosis/drug effects , Astrocytes/drug effects , Astrocytes/enzymology , Azoxymethane , Brain/enzymology , Brain/pathology , Brain Edema/chemically induced , Brain Edema/enzymology , Brain Edema/pathology , Cells, Cultured , Disease Models, Animal , Endothelial Growth Factors/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hepatic Encephalopathy/enzymology , Hepatic Encephalopathy/prevention & control , Immunohistochemistry , In Situ Nick-End Labeling , Interferon-gamma/metabolism , Lipopolysaccharides/pharmacology , Liver/enzymology , Liver/pathology , Liver Failure, Acute/chemically induced , Liver Failure, Acute/enzymology , Liver Failure, Acute/pathology , Macrophages/drug effects , Macrophages/enzymology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Peptides, Cyclic/pharmacology , Recombinant Fusion Proteins/metabolism , Time Factors , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/genetics , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL