Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Hepatol ; 66(5): 987-1000, 2017 05.
Article in English | MEDLINE | ID: mdl-28027971

ABSTRACT

BACKGROUND & AIMS: Hepatocyte transplantation partially corrects genetic disorders and has been associated anecdotally with reversal of acute liver failure. Monitoring for graft function and rejection has been difficult, and has contributed to limited graft survival. Here we aimed to use preparative liver-directed radiation therapy, and continuous monitoring for possible rejection in an attempt to overcome these limitations. METHODS: Preparative hepatic irradiation was examined in non-human primates as a strategy to improve engraftment of donor hepatocytes, and was then applied in human subjects. T cell immune monitoring was also examined in human subjects to assess adequacy of immunosuppression. RESULTS: Porcine hepatocyte transplants engrafted and expanded to comprise up to 15% of irradiated segments in immunosuppressed monkeys preconditioned with 10Gy liver-directed irradiation. Two patients with urea cycle deficiencies had early graft loss following hepatocyte transplantation; retrospective immune monitoring suggested the need for additional immunosuppression. Preparative radiation, anti-lymphocyte induction, and frequent immune monitoring were instituted for hepatocyte transplantation in a 27year old female with classical phenylketonuria. Post-transplant liver biopsies demonstrated multiple small clusters of transplanted cells, multiple mitoses, and Ki67+ hepatocytes. Mean peripheral blood phenylalanine (PHE) level fell from pre-transplant levels of 1343±48µM (normal 30-119µM) to 854±25µM (treatment goal ≤360µM) after transplant (36% decrease; p<0.0001), despite transplantation of only half the target number of donor hepatocytes. PHE levels remained below 900µM during supervised follow-up, but graft loss occurred after follow-up became inconsistent. CONCLUSIONS: Radiation preconditioning and serial rejection risk assessment may produce better engraftment and long-term survival of transplanted hepatocytes. Hepatocyte xenografts engraft for a period of months in non-human primates and may provide effective therapy for patients with acute liver failure. LAY SUMMARY: Hepatocyte transplantation can potentially be used to treat genetic liver disorders but its application in clinical practice has been impeded by inefficient hepatocyte engraftment and the inability to monitor rejection of transplanted liver cells. In this study, we first show in non-human primates that pretreatment of the host liver with radiation improves the engraftment of transplanted liver cells. We then used this knowledge in a series of clinical hepatocyte transplants in patients with genetic liver disorders to show that radiation pretreatment and rejection risk monitoring are safe and, if optimized, could improve engraftment and long-term survival of transplanted hepatocytes in patients.


Subject(s)
Graft Rejection , Hepatocytes/transplantation , Liver/radiation effects , Transplantation Conditioning , Adult , Animals , Female , Humans , Liver Diseases/therapy , Macaca fascicularis , Male , Swine , Transplantation, Heterologous
2.
Curr Protoc Toxicol ; 67: 14.13.1-14.13.27, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26828329

ABSTRACT

Liver disease is a major global health concern. Liver cirrhosis is one of the leading causes of death in the world and currently the only therapeutic option for end-stage liver disease (e.g., acute liver failure, cirrhosis, chronic hepatitis, cholestatic diseases, metabolic diseases, and malignant neoplasms) is orthotropic liver transplantation. Transplantation of hepatocytes has been proposed and used as an alternative to whole organ transplant to stabilize and prolong the lives of patients in some clinical cases. Although these experimental therapies have demonstrated promising and beneficial results, their routine use remains a challenge due to the shortage of donor livers available for cell isolation, variable quality of those tissues, the potential need for lifelong immunosuppression in the transplant recipient, and high costs. Therefore, new therapeutic strategies and more reliable clinical treatments are urgently needed. Recent and continuous technological advances in the development of stem cells suggest they may be beneficial in this respect. In this review, we summarize the history of stem cell and induced pluripotent stem cell (iPSC) technology in the context of hepatic differentiation and discuss the potential applications the technology may offer for human liver disease modeling and treatment. This includes developing safer drugs and cell-based therapies to improve the outcomes of patients with currently incurable health illnesses. We also review promising advances in other disease areas to highlight how the stem cell technology could be applied to liver diseases in the future. © 2016 by John Wiley & Sons, Inc.


Subject(s)
Hepatocytes/transplantation , Induced Pluripotent Stem Cells , Liver Diseases , Liver Regeneration , Regenerative Medicine/methods , Stem Cell Transplantation , Adult Stem Cells/cytology , Adult Stem Cells/transplantation , Animals , Cell Differentiation , Disease Models, Animal , Hepatocytes/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Liver Diseases/etiology , Liver Diseases/therapy
3.
Drug Metab Dispos ; 44(7): 1027-37, 2016 07.
Article in English | MEDLINE | ID: mdl-26921389

ABSTRACT

Variability in drug-metabolizing enzyme developmental trajectories contributes to interindividual differences in susceptibility to chemical toxicity and adverse drug reactions, particularly in the first years of life. Factors linked to these interindividual differences are largely unknown, but molecular mechanisms regulating ontogeny are likely involved. To evaluate chromatin structure dynamics as a likely contributing mechanism, age-dependent changes in modified and variant histone occupancy were evaluated within known CYP3A4 and 3A7 regulatory domains. Chromatin immunoprecipitation using fetal or postnatal human hepatocyte chromatin pools followed by quantitative polymerase chain reaction DNA amplification was used to determine relative chromatin occupancy by modified and variant histones. Chromatin structure representing a poised transcriptional state (bivalent chromatin), indicated by the occupancy by modified histones associated with both active and repressed transcription, was observed for CYP3A4 and most 3A7 regulatory regions in both postnatal and fetal livers. However, the CYP3A4 regulatory regions had significantly greater occupancy by modified histones associated with repressed transcription in the fetal liver. Conversely, some modified histones associated with active transcription exhibited greater occupancy in the postnatal liver. CYP3A7 regulatory regions also had significantly greater occupancy by modified histones associated with repressed transcription in the fetus. The observed occupancy by modified histones is consistent with chromatin structural dynamics contributing to CYP3A4 ontogeny, although the data are less conclusive regarding CYP3A7. Interpretation of the latter data may be confounded by cell-type heterogeneity in the fetal liver.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/metabolism , Cytochrome P-450 CYP3A/metabolism , Histones/metabolism , Liver/enzymology , Adult , Age Factors , Aged , Aged, 80 and over , Binding Sites , Child , Child, Preschool , Chromatin/chemistry , Chromatin/genetics , Cytochrome P-450 CYP3A/genetics , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Gestational Age , Hepatocytes/enzymology , Histones/chemistry , Histones/genetics , Humans , Infant , Liver/embryology , Middle Aged , Nucleic Acid Conformation , Promoter Regions, Genetic , Protein Conformation , Structure-Activity Relationship , Transcription, Genetic
4.
Oncotarget ; 7(14): 17681-98, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-26769846

ABSTRACT

Chronic alcohol exposure increased hepatic receptor-interacting protein kinase (RIP) 3 expression and necroptosis in the liver but its mechanisms are unclear. In the present study, we demonstrated that chronic alcohol feeding plus binge (Gao-binge) increased RIP3 but not RIP1 protein levels in mouse livers. RIP3 knockout mice had decreased serum alanine amino transferase activity and hepatic steatosis but had no effect on hepatic neutrophil infiltration compared with wild type mice after Gao-binge alcohol treatment. The hepatic mRNA levels of RIP3 did not change between Gao-binge and control mice, suggesting that alcohol-induced hepatic RIP3 proteins are regulated at the posttranslational level. We found that Gao-binge treatment decreased the levels of proteasome subunit alpha type-2 (PSMA2) and proteasome 26S subunit, ATPase 1 (PSMC1) and impaired hepatic proteasome function. Pharmacological or genetic inhibition of proteasome resulted in the accumulation of RIP3 in mouse livers. More importantly, human alcoholics had decreased expression of PSMA2 and PSMC1 but increased protein levels of RIP3 compared with healthy human livers. Moreover, pharmacological inhibition of RIP1 decreased Gao-binge-induced hepatic inflammation, neutrophil infiltration and NF-κB subunit (p65) nuclear translocation but failed to protect against steatosis and liver injury induced by Gao-binge alcohol. In conclusion, results from this study suggest that impaired hepatic proteasome function by alcohol exposure may contribute to hepatic accumulation of RIP3 resulting in necroptosis and steatosis while RIP1 kinase activity is important for alcohol-induced inflammation.


Subject(s)
Fatty Liver/enzymology , Liver Diseases, Alcoholic/enzymology , Receptor-Interacting Protein Serine-Threonine Kinases/biosynthesis , Animals , Binge Drinking/enzymology , Binge Drinking/pathology , Ethanol/administration & dosage , GTPase-Activating Proteins/biosynthesis , GTPase-Activating Proteins/metabolism , Humans , Liver/drug effects , Liver/enzymology , Liver/pathology , Liver Diseases, Alcoholic/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Pore Complex Proteins/biosynthesis , Nuclear Pore Complex Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
5.
Mol Biosyst ; 12(2): 566-74, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26672496

ABSTRACT

Methylmalonic acidemia (MMA) is a heterogeneous and severe autosomal recessive inborn error of metabolism most commonly caused by the deficient activity of the vitamin B12 dependent enzyme, methylmalonyl-CoA mutase (MUT). The main treatment for MMA patients is the dietary restriction of propiogenic amino acids and carnitine supplementation. Despite treatment, the prognosis for vitamin B12 non-responsive patients remains poor and is associated with neonatal lethality, persistent morbidity and decreased life expectancy. While multi-organ pathology is a feature of MMA, the liver is severely impacted by mitochondrial dysfunction which likely underlies the metabolic instability experienced by the patients. Liver and/or combined liver/kidney transplantation is therefore sometimes performed in severely affected patients. Using liver specimens from donors and MMA patients undergoing elective liver transplantation collected under a dedicated natural history protocol (clinicaltrials.gov: NCT00078078), we employed proteomics to characterize the liver pathology and impaired hepatic metabolism observed in the patients. Pathway analysis revealed perturbations of enzymes involved in energy metabolism, gluconeogenesis and Krebs cycle anaplerosis. Our findings identify new pathophysiologic and therapeutic targets that could be valuable for designing alternative therapies to alleviate clinical manifestations seen in this disorder.


Subject(s)
Amino Acid Metabolism, Inborn Errors/metabolism , Liver/metabolism , Proteome/metabolism , Amino Acid Metabolism, Inborn Errors/surgery , Case-Control Studies , Child, Preschool , Female , Humans , Infant , Liver Transplantation , Male , Metabolic Networks and Pathways , Two-Dimensional Difference Gel Electrophoresis
6.
Toxicol Appl Pharmacol ; 289(2): 213-22, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26431796

ABSTRACT

3'-Hydroxyacetanilide orN-acetyl-meta-aminophenol (AMAP) is generally regarded as a non-hepatotoxic analog of acetaminophen (APAP). Previous studies demonstrated the absence of toxicity after AMAP in mice, hamsters, primary mouse hepatocytes and several cell lines. In contrast, experiments with liver slices suggested that it may be toxic to human hepatocytes; however, the mechanism of toxicity is unclear. To explore this,we treated primary human hepatocytes (PHH) with AMAP or APAP for up to 48 h and measured several parameters to assess metabolism and injury. Although less toxic than APAP, AMAP dose-dependently triggered cell death in PHH as indicated by alanine aminotransferase (ALT) release and propidium iodide (PI) staining. Similar to APAP, AMAP also significantly depleted glutathione (GSH) in PHH and caused mitochondrial damage as indicated by glutamate dehydrogenase (GDH) release and the JC-1 assay. However, unlike APAP, AMAP treatment did not cause relevant c-jun-N-terminal kinase (JNK) activation in the cytosol or phospho-JNK translocation to mitochondria. To compare, AMAP toxicity was assessed in primary mouse hepatocytes (PMH). No cytotoxicity was observed as indicated by the lack of lactate dehydrogenase release and no PI staining. Furthermore, there was no GSH depletion or mitochondrial dysfunction after AMAP treatment in PMH. Immunoblotting for arylated proteins suggested that AMAP treatment caused extensive mitochondrial protein adduct formation in PHH but not in PMH. In conclusion, AMAP is hepatotoxic in PHH and the mechanism involves the formation of mitochondrial protein adducts and mitochondrial dysfunction.


Subject(s)
Acetanilides/toxicity , Chemical and Drug Induced Liver Injury/etiology , Hepatocytes/drug effects , Liver/drug effects , Mitochondria, Liver/drug effects , Mitochondrial Proteins/metabolism , Animals , Cell Death/drug effects , Cells, Cultured , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Dose-Response Relationship, Drug , Glutamate Dehydrogenase/metabolism , Glutathione/metabolism , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , L-Lactate Dehydrogenase/metabolism , Liver/metabolism , Liver/pathology , Mice , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Phosphorylation , Primary Cell Culture , Protein Binding , Signal Transduction/drug effects , Species Specificity , Time Factors
7.
J Biol Chem ; 290(36): 22236-49, 2015 Sep 04.
Article in English | MEDLINE | ID: mdl-26221041

ABSTRACT

Arginine methylation is a common post-translational modification, but its role in regulating protein function is poorly understood. This study demonstrates that, TNF receptor-associated factor 6 (TRAF6), an E3 ubiquitin ligase involved in innate immune signaling, is regulated by reversible arginine methylation in a range of primary and cultured cells. Under basal conditions, TRAF6 is methylated by the methyltransferase PRMT1, and this inhibits its ubiquitin ligase activity, reducing activation of toll-like receptor signaling. In response to toll-like receptor ligands, TRAF6 is demethylated by the Jumonji domain protein JMJD6. Demethylation is required for maximal activation of NF-κB. Loss of JMJD6 leads to reduced response, and loss of PRMT1 leads to basal pathway activation with subsequent desensitization to ligands. In human primary cells, variations in the PRMT1/JMJD6 ratio significantly correlate with TRAF6 methylation, basal activation of NF-κB, and magnitude of response to LPS. Reversible arginine methylation of TRAF6 by the opposing effects of PRMT1 and JMJD6 is, therefore, a novel mechanism for regulation of innate immune pathways.


Subject(s)
Arginine/metabolism , Signal Transduction , TNF Receptor-Associated Factor 6/metabolism , Toll-Like Receptors/metabolism , Animals , Blotting, Western , Cell Line , Cell Line, Tumor , Cells, Cultured , Humans , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Kinetics , Ligands , Male , Methylation , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , RNA Interference , Repressor Proteins/genetics , Repressor Proteins/metabolism , TNF Receptor-Associated Factor 6/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
8.
Toxicol Appl Pharmacol ; 283(3): 168-77, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25636263

ABSTRACT

Accumulation of bile acids is a major mediator of cholestatic liver injury. Recent studies indicate bile acid composition between humans and rodents is dramatically different, as humans have a higher percent of glycine conjugated bile acids and increased chenodeoxycholate content, which increases the hydrophobicity index of bile acids. This increase may lead to direct toxicity that kills hepatocytes, and promotes inflammation. To address this issue, this study assessed how pathophysiological concentrations of bile acids measured in cholestatic patients affected primary human hepatocytes. Individual bile acid levels were determined in serum and bile by UPLC/QTOFMS in patients with extrahepatic cholestasis with, or without, concurrent increases in serum transaminases. Bile acid levels increased in serum of patients with liver injury, while biliary levels decreased, implicating infarction of the biliary tracts. To assess bile acid-induced toxicity in man, primary human hepatocytes were treated with relevant concentrations, derived from patient data, of the model bile acid glycochenodeoxycholic acid (GCDC). Treatment with GCDC resulted in necrosis with no increase in apoptotic parameters. This was recapitulated by treatment with biliary bile acid concentrations, but not serum concentrations. Marked elevations in serum full-length cytokeratin-18, high mobility group box 1 protein (HMGB1), and acetylated HMGB1 confirmed inflammatory necrosis in injured patients; only modest elevations in caspase-cleaved cytokeratin-18 were observed. These data suggest human hepatocytes are more resistant to human-relevant bile acids than rodent hepatocytes, and die through necrosis when exposed to bile acids. These mechanisms of cholestasis in humans are fundamentally different to mechanisms observed in rodent models.


Subject(s)
Bile Acids and Salts/toxicity , Cholestasis, Extrahepatic/pathology , Glycochenodeoxycholic Acid/toxicity , Hepatocytes/drug effects , Jaundice, Obstructive/pathology , Acetylation , Animals , Bile Acids and Salts/blood , Biomarkers/blood , Cells, Cultured , Cholestasis, Extrahepatic/blood , Dose-Response Relationship, Drug , HMGB1 Protein/blood , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Jaundice, Obstructive/blood , Keratin-18/blood , Mice, Inbred C57BL , Necrosis , Primary Cell Culture , Species Specificity
9.
J Biol Chem ; 290(12): 7871-86, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25623066

ABSTRACT

Although endogenous mechanisms that negatively regulate cytochrome P450 (P450) monooxygenases in response to physiological and pathophysiological signals are not well understood, they are thought to result from alterations in the level of endogenous metabolites, involved in maintaining homeostasis. Here we show that homeostatic changes in hepatic metabolite profile in Abcb6 (mitochondrial ATP-binding cassette transporter B6) deficiency results in suppression of a specific subset of hepatic P450 activity. Abcb6 null mice are more susceptible to pentobarbital-induced sleep and zoxazolamine-induced paralysis, secondary to decreased expression and activity of Cyp3a11 and Cyp2b10. The knock-out mice also show decrease in both basal and xeno-inducible expression and activity of a subset of hepatic P450s that appear to be related to changes in hepatic metabolite profile. These data, together with the observation that liver extracts from Abcb6-deficient mice suppress P450 expression in human primary hepatocytes, suggest that this mouse model may provide an opportunity to understand the physiological signals and the mechanisms involved in negative regulation of P450s.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Cytochrome P-450 Enzyme System/metabolism , Liver/metabolism , Animals , Base Sequence , DNA Primers , Liver/enzymology , Mass Spectrometry , Mice
10.
J Clin Transl Res ; 1(2): 100-106, 2015.
Article in English | MEDLINE | ID: mdl-26722668

ABSTRACT

BACKGROUND: Acetaminophen (APAP) hepatotoxicity is a major cause of acute liver failure in many countries. Mechanistic studies in mice and humans have implicated formation of a reactive metabolite, mitochondrial dysfunction and oxidant stress as critical events in the pathophysiology of APAP-induced liver cell death. It was recently suggested that ATP released from necrotic cells can directly cause cell death in mouse hepatocytes and in a hepatoma cell line (HepG2). AIM: To assess if ATP can directly cause cell toxicity in hepatocytes and evaluate their relevance in the human system. METHODS: Primary mouse hepatocytes, human HepG2 cells, the metabolically competent human HepaRG cell line and freshly isolated primary human hepatocytes were exposed to 10-100 µM ATP or ATγP in the presence or absence of 5-10 mM APAP for 9-24 h. RESULTS: ATP or ATγP was unable to directly cause cell toxicity in all 4 types of hepatocytes. In addition, ATP did not enhance APAP-induced cell death observed in primary mouse or human hepatocytes, or in HepaRG cells as measured by LDH release and by propidium iodide staining in primary mouse hepatocytes. Furthermore, addition of ATP did not cause mitochondrial dysfunction or enhance APAP-induced mitochondrial dysfunction in primary murine hepatocytes, although ATP did cause cell death in murine RAW macrophages. CONCLUSIONS: It is unlikely that ATP released from necrotic cells can significantly affect cell death in human or mouse liver during APAP hepatotoxicity. RELEVANCE FOR PATIENTS: Understanding the mechanisms of APAP-induced cell injury is critical for identifying novel therapeutic targets to prevent liver injury and acute liver failure in APAP overdose patients.

11.
J Clin Transl Res ; 1(2): 1-7, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-30873447

ABSTRACT

BACKGROUND: Acetaminophen (APAP) hepatotoxicity is a major cause of acute liver failure in many countries. Mechanistic studies in mice and humans have implicated formation of a reactive metabolite, mitochondrial dysfunction and oxidant stress as critical events in the pathophysiology of APAP-induced liver cell death. It was recently suggested that ATP released from necrotic cells can directly cause cell death in mouse hepatocytes and in a hepatoma cell line (HepG2). AIM: To assess if ATP can directly cause cell toxicity in hepatocytes and evaluate their relevance in the human system. METHODS: Primary mouse hepatocytes, human HepG2 cells, the metabolically competent human HepaRG cell line and freshly isolated primary human hepatocytes were exposed to 10-100 µM ATP or ATγPin the presence or absence of 5-10 mM APAP for 9-24 h. RESULTS: ATP or ATγP was unable to directly cause cell toxicity in all 4 types of hepatocytes. In addition, ATP did not enhance APAP-induced cell death observed in primary mouse or human hepatocytes, or in HepaRG cells as measured by LDH release and by propidium iodide staining in primary mouse hepatocytes. Furthermore, addition of ATP did not cause mitochondrial dysfunction or enhance APAP-induced mitochondrial dysfunction in primary murine hepatocytes, although ATP did cause cell death in murine RAW macrophages. CONCLUSIONS: It is unlikely that ATP released from necrotic cells can significantly affect cell death in human or mouse liver during APAP hepatotoxicity. RELEVANCE FOR PATIENTS: Understanding the mechanisms of APAP-induced cell injury is critical for identifying novel therapeutic targets to prevent liver injury and acute liver failure in APAP overdose patients.

12.
Curr Protoc Toxicol ; 62: 14.12.1-23, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25378242

ABSTRACT

Orthotopic liver transplantation remains the only curative treatment for many end-stage liver diseases, yet the number of patients receiving liver transplants remains limited by the number of organs available for transplant. There is a need for alternative therapies for liver diseases. The transplantation of isolated hepatocytes (liver cells) has been used as an experimental therapy for liver disease in a limited number of cases. Recently, the 100th case of hepatocyte transplantation was reported. This review discusses the history of the hepatocyte transplant field, the major discoveries that supported and enabled the first hepatocyte transplants, and reviews the cases and outcomes of the first 100 clinical transplants. Some of the problems that limit the application or efficacy of hepatocyte transplantation are discussed, as are possible solutions to these problems. In conclusion, hepatocyte transplants have proven effective particularly in cases of metabolic liver disease where reversal or amelioration of the characteristic symptoms of the disease is easily quantified. However, no patients have been completely corrected of a metabolic liver disease for a significant amount of time by hepatocyte transplantation alone. It is likely that future developments in new sources of cells for transplantation will be required before this cellular therapy can be fully implemented and available for large numbers of patients.


Subject(s)
Hepatocytes/cytology , Liver Diseases/therapy , Humans , Liver Diseases/pathology , Liver Transplantation
13.
Toxicol Appl Pharmacol ; 279(3): 266-274, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24905542

ABSTRACT

UNLABELLED: Acetaminophen (APAP) overdose is the most prevalent cause of drug-induced liver injury in western countries. Numerous studies have been conducted to investigate the mechanisms of injury after APAP overdose in various animal models; however, the importance of these mechanisms for humans remains unclear. Here we investigated APAP hepatotoxicity using freshly isolated primary human hepatocytes (PHH) from either donor livers or liver resections. PHH were exposed to 5mM, 10mM or 20mM APAP over a period of 48 h and multiple parameters were assessed. APAP dose-dependently induced significant hepatocyte necrosis starting from 24h, which correlated with the clinical onset of human liver injury after APAP overdose. Interestingly, cellular glutathione was depleted rapidly during the first 3h. APAP also resulted in early formation of APAP-protein adducts (measured in whole cell lysate and in mitochondria) and mitochondrial dysfunction, indicated by the loss of mitochondrial membrane potential after 12h. Furthermore, APAP time-dependently triggered c-Jun N-terminal kinase (JNK) activation in the cytosol and translocation of phospho-JNK to the mitochondria. Both co-treatment and post-treatment (3h) with the JNK inhibitor SP600125 reduced JNK activation and significantly attenuated cell death at 24h and 48h after APAP. The clinical antidote N-acetylcysteine offered almost complete protection even if administered 6h after APAP and a partial protection when given at 15 h. CONCLUSION: These data highlight important mechanistic events in APAP toxicity in PHH and indicate a critical role of JNK in the progression of injury after APAP in humans. The JNK pathway may represent a therapeutic target in the clinic.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Cell Death/drug effects , Hepatocytes/drug effects , Acetaminophen/antagonists & inhibitors , Acetylcysteine/pharmacology , Adult , Aged , Antidotes/pharmacology , Enzyme Activation/drug effects , Female , Glutathione/metabolism , Hepatocytes/enzymology , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Middle Aged , Mitochondria, Liver/drug effects , Mitochondrial Diseases/chemically induced , Mitochondrial Diseases/metabolism , Necrosis/pathology , Primary Cell Culture , Proteins/metabolism , Subcellular Fractions/drug effects , Subcellular Fractions/enzymology , Subcellular Fractions/metabolism , Young Adult
14.
Cell Transplant ; 23(12): 1545-56, 2014.
Article in English | MEDLINE | ID: mdl-24702711

ABSTRACT

Transplantation of human hepatocytes (HTx) has gained recognition as a bridge to, or an alternative to, orthotopic liver transplantation for patients with acute liver failure or genetic defects in liver function. Although the quality of the hepatocytes used for cell transplantation is critical, no consensus exists on protocols to assess the function of hepatocytes prior to HTx. Application of this cell therapy in clinical practice could be aided by fast and reliable assays to evaluate the functional competence of isolated hepatocytes prior to clinical transplantation. Traditional assays for measuring metabolic functions in primary hepatocytes frequently involve highly technical equipment, time-consuming methods, and large numbers of cells. We describe a novel approach for the rapid assessment of the metabolic capabilities of human hepatocytes. This report details simple procedures to evaluate 11 endpoints from cells isolated from human liver that can be performed by a single operator within approximately 2 h of isolation. Longer term cultured hepatocytes were also analyzed to determine if the results from the 2-h tests were predictive of long-term hepatic function. The assays simultaneously measure five cytochrome P450 activities, one phase II activity, plating efficiency, and ammonia metabolism in addition to viability and cell yield. The assays require fewer than 20 million cells and can be completed using commonly available and inexpensive laboratory equipment. The protocol details methods that can be used in a time frame that would allow analysis of hepatic functions in freshly isolated hepatocytes prior to their use for clinical transplantation.


Subject(s)
Biological Assay/methods , Hepatocytes/cytology , Hepatocytes/metabolism , Adolescent , Adult , Aged , Ammonia/metabolism , Caspases/metabolism , Cell Separation , Cell Survival , Child , Child, Preschool , Cytochrome P-450 Enzyme System/metabolism , Female , Humans , Infant , Liver Transplantation , Male , Middle Aged , Young Adult
15.
Drug Metab Dispos ; 42(5): 863-6, 2014 May.
Article in English | MEDLINE | ID: mdl-24513655

ABSTRACT

Bedaquiline is a recently approved drug for the treatment of multidrug-resistant tuberculosis. Adverse cardiac and hepatic drug reactions to bedaquiline have been noted in clinical practice. The current study investigated bedaquiline metabolism in human hepatocytes using a metabolomic approach. Bedaquiline N-demethylation via CYP3A4 was confirmed as the major pathway in bedaquiline metabolism. In addition to CYP3A4, we found that both CYP2C8 and CYP2C19 contributed to bedaquiline N-demethylation. The Km values of CYP2C8, CYP2C19, and CYP3A4 in bedaquiline N-demethylation were 13.1, 21.3, and 8.5 µM, respectively. We also identified a novel metabolic pathway of bedaquiline that produced an aldehyde intermediate. In summary, this study extended our knowledge of bedaquiline metabolism, which can be applied to predict and prevent drug-drug interactions and adverse drug reactions associated with bedaquiline.


Subject(s)
Antitubercular Agents/metabolism , Aryl Hydrocarbon Hydroxylases/metabolism , Cytochrome P-450 CYP2C19/metabolism , Cytochrome P-450 CYP2C8/metabolism , Cytochrome P-450 CYP3A/metabolism , Diarylquinolines/metabolism , Hepatocytes/metabolism , Antitubercular Agents/pharmacokinetics , Aryl Hydrocarbon Hydroxylases/genetics , Cells, Cultured , Cytochrome P-450 CYP2C19/genetics , Cytochrome P-450 CYP2C8/genetics , Cytochrome P-450 CYP3A/genetics , Dealkylation , Diarylquinolines/pharmacokinetics , Hepatocytes/drug effects , Hepatocytes/enzymology , Humans , Metabolomics , Methylation , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Tuberculosis, Multidrug-Resistant/drug therapy , Tuberculosis, Multidrug-Resistant/enzymology , Tuberculosis, Multidrug-Resistant/metabolism
16.
Cell Transplant ; 23(9): 1143-51, 2014.
Article in English | MEDLINE | ID: mdl-23768881

ABSTRACT

Transplantation of human hepatocytes is gaining recognition as a bridge or an alternative to orthotopic liver transplantation for patients with acute liver failure and genetic defects. Since most patients require multiple cell infusions over an extended period of time, we investigated hepatic functions in cells maintained in University of Wisconsin solution at 4°C up to 72 h. Eleven different assessments of hepatic viability and function were investigated both pre- and posthypothermic storage, including plating efficiency, caspase-3/7 activity, ammonia metabolism, and drug-metabolizing capacity of isolated hepatocytes. Long-term function, basal, and induced cytochrome P450 activities were measured after exposure to prototypical inducing agents. Cells from 47 different human liver specimens were analyzed. Viability significantly decreased in cells cold stored in UW solution, while apoptosis level and plating efficiency were not significantly different from fresh cells. Luminescent and fluorescent methods assessed phases I and II activities both pre- and post-24-72 h of cold preservation. A robust induction (up to 200-fold) of phase I enzymes was observed in cultured cells. Phase II and ammonia metabolism remained stable during hypothermic storage, although the inductive effect of culture on each metabolic activity was eventually lost. Using techniques that characterize 11 measurements of hepatic viability and function from plating efficiency, to ammonia metabolism, to phases I and II drug metabolism, it was determined that while viability decreased, the remaining viable cells in cold-stored suspensions retained critical hepatic functions for up to 48 h at levels not significantly different from those observed in freshly isolated cells.


Subject(s)
Cryopreservation/methods , Hepatocytes/cytology , Adenosine/chemistry , Adenosine/pharmacology , Adolescent , Adult , Aged , Allopurinol/chemistry , Allopurinol/pharmacology , Ammonia/metabolism , Caspase 3/metabolism , Caspase 7/metabolism , Cell Survival/drug effects , Cells, Cultured , Child , Child, Preschool , Cold Temperature , Cytochrome P-450 Enzyme System/metabolism , Female , Glutathione/chemistry , Glutathione/pharmacology , Hepatocytes/metabolism , Hepatocytes/transplantation , Humans , Infant , Insulin/chemistry , Insulin/pharmacology , Male , Middle Aged , Organ Preservation Solutions/chemistry , Organ Preservation Solutions/pharmacology , Raffinose/chemistry , Raffinose/pharmacology , Young Adult
17.
Cell Transplant ; 23(1): 27-38, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23394081

ABSTRACT

Hepatocyte transplantation has been used to treat liver disease. The availability of cells for these procedures is quite limited. Human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs) may be a useful source of hepatocytes for basic research and transplantation if efficient and effective differentiation protocols were developed and problems with tumorigenicity could be overcome. Recent evidence suggests that the cell of origin may affect hiPSC differentiation. Thus, hiPSCs generated from hepatocytes may differentiate back to hepatocytes more efficiently than hiPSCs from other cell types. We examined the efficiency of reprogramming adult and fetal human hepatocytes. The present studies report the generation of 40 hiPSC lines from primary human hepatocytes under feeder-free conditions. Of these, 37 hiPSC lines were generated from fetal hepatocytes, 2 hiPSC lines from normal hepatocytes, and 1 hiPSC line from hepatocytes of a patient with Crigler-Najjar syndrome, type 1. All lines were confirmed reprogrammed and expressed markers of pluripotency by gene expression, flow cytometry, immunocytochemistry, and teratoma formation. Fetal hepatocytes were reprogrammed at a frequency over 50-fold higher than adult hepatocytes. Adult hepatocytes were only reprogrammed with six factors, while fetal hepatocytes could be reprogrammed with three (OCT4, SOX2, NANOG) or four factors (OCT4, SOX2, NANOG, LIN28 or OCT4, SOX2, KLF4, C-MYC). The increased reprogramming efficiency of fetal cells was not due to increased transduction efficiency or vector toxicity. These studies confirm that hiPSCs can be generated from adult and fetal hepatocytes including those with genetic diseases. Fetal hepatocytes reprogram much more efficiently than adult hepatocytes, although both could serve as useful sources of hiPSC-derived hepatocytes for basic research or transplantation.


Subject(s)
Cellular Reprogramming/physiology , Embryonic Stem Cells/physiology , Hepatocytes/physiology , Induced Pluripotent Stem Cells/physiology , Adult , Animals , Cell Culture Techniques , Cell Differentiation/physiology , Cell Growth Processes/physiology , Cells, Cultured , Embryonic Stem Cells/cytology , Female , Hepatocytes/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Infant , Infant, Newborn , Kruppel-Like Factor 4 , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Young Adult
18.
Stem Cell Res ; 11(1): 563-73, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23644508

ABSTRACT

UNLABELLED: Domino liver transplantation is a method used to increase the number of liver grafts available for orthotopic liver transplantation (OLT). Reports indicate that livers from patients with metabolic liver disease can be safely transplanted into select recipients if the donor's defect and the recipient's metabolic needs are carefully considered. The liver of patients with many types of metabolic liver disease is morphologically and biochemically normal, except for the mutation that characterizes that disease. Other biochemical functions normally performed by the liver are present and presumably "normal" in these hepatocytes. Hepatocytes were isolated from the liver of 35 organ donors and 35 liver tissues taken at OLT from patients with liver disease were analyzed for 9 different measures of viability and function. The data indicate that cells isolated from some diseased livers performed as well or better than those isolated from organ donors with respect to viability, cell yield, plating efficiency and in assays of liver function, including drug metabolism, conjugation reactions and ammonia metabolism. Cells from metabolic diseased livers rapidly and efficiently repopulated a mouse liver upon transplantation. CONCLUSIONS: As with domino liver transplantation, domino cell transplantation deserves consideration as method to extend the pool of available organs and cells for transplantation.


Subject(s)
Hepatocytes/transplantation , Liver Diseases/pathology , Liver Transplantation/methods , Liver/pathology , Adolescent , Adult , Animals , Child , Child, Preschool , Female , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Infant , Liver/metabolism , Liver/surgery , Liver Diseases/metabolism , Liver Diseases/surgery , Male , Mice , Young Adult
19.
Mol Genet Metab ; 109(2): 132-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23566440

ABSTRACT

Orthotopic liver transplant (OLT) significantly improves patient outcomes in maple syrup urine disease (MSUD; OMIM: 248600), yet organ shortages point to the need for alternative therapies. Hepatocyte transplantation has shown both clinical and preclinical efficacy as an intervention for metabolic liver diseases, yet the availability of suitable livers for hepatocyte isolation is also limited. Conversely, human amnion epithelial cells (hAEC) may have utility as a hepatocyte substitute, and they share many of the characteristics of pluripotent embryonic stem cells while lacking their safety and ethical concerns. We reported that like hepatocytes, transplantation of hAEC significantly improved survival and lifespan, normalized body weight, and significantly improved branched-chain amino acid (BCAA) levels in sera and brain in a transgenic murine model of intermediate maple syrup urine disease (imsud). In the current report, we detail the neural and peripheral metabolic improvements associated with hAEC transplant in imsud mice, including amino acids associated with bioenergetics, the urea cycle, as well as the neurotransmitter systems for serotonin, dopamine, and gamma-aminobutyric acid (GABA). This stem cell therapy results in significant global correction of the metabolic profile that characterizes the disease, both in the periphery and the central nervous system, the target organ for toxicity in iMSUD. The significant correction of the disease phenotype, coupled with the theoretical benefits of hAEC, particularly their lack of immunogenicity and tumorigenicity, suggests that human amnion epithelial cells deserve serious consideration for clinical application to treat metabolic liver diseases.


Subject(s)
Amino Acids/blood , Amnion/cytology , Epithelial Cells/transplantation , Maple Syrup Urine Disease/therapy , Neurotransmitter Agents/metabolism , Animals , Brain/metabolism , Citric Acid Cycle , Humans , Maple Syrup Urine Disease/blood , Mice , Mice, Transgenic
20.
Drug Metab Dispos ; 41(2): 296-304, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23129211

ABSTRACT

Little information is available in the literature regarding the expression and activity of transporters in fetal human liver or cultured cells. A synthetic progesterone structural analog, 17α-hydroxyprogesterone caproate (17-OHPC), is used in the prevention of spontaneous abortion in women with a history of recurrent miscarriage (habitual abortion). 17-OHPC has been reported to traverse the placental barrier and gain access to fetal circulation. In this study, the role of transporters in the disposition of 17-OHPC in fetal and adult human hepatocytes was examined. Progesterone metabolites have been reported to induce trans-inhibition of bile acid transporter, ABCB11. Thus, we investigated the effect of 17-OHPC or its metabolites on [(3)H]taurocholic acid transport in sandwich-cultured human fetal and adult hepatocytes. 17-OHPC was taken up rapidly into the cells and transported out partially by an active efflux process that was significantly inhibited by cold temperature, cyclosporine, verapamil, and rifampin. The active efflux mechanism was observed in both adult and fetal hepatocyte cultures. 17-OHPC produced a concentration-dependent inhibition of taurocholate efflux into canaliculi in sandwich-cultured adult and fetal human hepatocytes. However, given the high concentrations required to cause inhibition of these transport processes, no adverse effects would be anticipated from therapeutic levels of 17-OHPC. We also evaluated the expression of various hepatic transporters (ABCB1, ABCB4, SLCO1B1, SLCO1B3, SLCO2B1, ABCB11, SLC10A1, ABCC2, ABCC3, ABCC4, and ABCG2) in fetal and adult hepatocytes. With the exception of ABCB4, all transporters examined were expressed, albeit at lower mRNA levels in fetal hepatocytes compared with adults.


Subject(s)
Hepatocytes/metabolism , Hydroxyprogesterones/metabolism , Membrane Transport Proteins/metabolism , Taurocholic Acid/metabolism , 17 alpha-Hydroxyprogesterone Caproate , Adult , Age Factors , Aged , Biological Transport , Cells, Cultured , Cold Temperature , Cyclosporine/pharmacology , Female , Gestational Age , Hepatocytes/drug effects , Humans , Hydroxyprogesterones/pharmacology , Kinetics , Male , Membrane Transport Proteins/drug effects , Membrane Transport Proteins/genetics , Middle Aged , Multidrug Resistance-Associated Protein 2 , RNA, Messenger/metabolism , Rifampin/pharmacology , Verapamil/pharmacology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...