1
|
Hu C, Shi J, Zhang F, Lv M, Ge Z, Feng M, Fan Z, Liu D, Du J, Sun Y. Ginsenoside Rd-Loaded Antioxidant Polymersomes to Regulate Mitochondrial Homeostasis for Bone Defect Healing in Periodontitis. Adv Healthc Mater 2025; 14:e2403817. [PMID: 39703116 DOI: 10.1002/adhm.202403817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/04/2024] [Indexed: 12/21/2024]
Abstract
Periodontitis is the leading cause of tooth loss in adults. Initially triggered by bacterial infection, it is characterized by subsequent dysregulation of mitochondrial homeostasis, leading to ongoing loss of periodontal tissue. Mitophagic flux, a critical physiological mechanism for maintaining mitochondrial homeostasis, is compromised in periodontitis. Additionally, increased release of reactive oxygen species (ROS) exacerbates mitochondrial damage. In this study, a ginsenoside Rd (Rd)-loaded antioxidative polymersome (RdAP) is designed, which is self-assembled from a mitochondrial-protective and ROS-scavenging block copolymer, poly(ethylene oxide)-block-poly(phenylboronic acid pinacol ester-conjugated polylysine) (PEO113-b-P(Lys-PAPE)60). The phenylboronic acid pinacol ester (PAPE) segment exhibits excellent ROS-responsive properties, enabling effective ROS scavenging through antioxidant production. Rd significantly enhances mitophagic flux by 2.5-fold in periodontal ligament stem cells (PDLSCs) under oxidative stress. Together with the antioxidative polymersome, RdAPs restore mitochondrial homeostasis and enhance the osteogenic capacity of PDLSCs, bringing it closer to that of healthy controls. In a mouse model of periodontitis, the bone mass in the RdAP-treated group is 1.37 times greater than that in the untreated periodontitis group. Overall, the findings propose a novel strategy for addressing refractory periodontitis, which may also be applicable to other diseases characterized by mitochondrial homeostasis imbalance.
Collapse
Affiliation(s)
- Congjiao Hu
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Implantology, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China
| | - Junqiu Shi
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai, 201804, China
| | - Fan Zhang
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Implantology, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China
| | - Mingchen Lv
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai, 201804, China
| | - Zhenghong Ge
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai, 201804, China
| | - Meiting Feng
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Implantology, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China
| | - Zhen Fan
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai, 201804, China
| | - Danqing Liu
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Jianzhong Du
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai, 201804, China
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
- School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Yao Sun
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Implantology, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China
| |
Collapse
|
2
|
Li X, Wang M, Zhong Y, Yin Q, Hu Z, Tian W, Liu Z, Liu Z. Comparative pharmacokinetics of six components in normal and rheumatoid arthritis rats after intragastrical administration of Qianghuo Shengshi Decoction granules by LC-MS/MS. CHINESE HERBAL MEDICINES 2024; 16:457-465. [PMID: 39072204 PMCID: PMC11283214 DOI: 10.1016/j.chmed.2023.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/13/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2024] Open
Abstract
Objective To investigate the plasma pharmacokinetics of six representative components (nodakenin, osthole, 5-O-methylvisammioside, ferulic acid, liquiritigenin, and liquiritin), which were the ingredients of Qianghuo Shengshi Decoction (QSD) granules, in normal and rheumatoid arthritis (RA) rats administrated QSD granules intragastrically. Methods A rapid and accurate ultra-high performance liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was developed for the simultaneous determination of six components in plasma, and it showed a good specificity, linearity, intra-day and inter-day precision, intra-day and inter-day accuracy, extraction recovery, stability, and the less matrix effect. Results The validated LC-MS/MS method was successfully used to compare the plasma pharmacokinetics of six ingredients between normal and RA rats after intragastrical administration of QSD granules and differences in the pharmacokinetics were found in two types of rats. The absorption rate in the RA rats was lower for nodakenin, osthole, 5-O-methylvisammioside, liquiritigenin and liquiritin than in the normal group, while the absorption rate of ferulic acid remained constant in two groups. In comparison with the normal rats, the exposure concentration of nodakenin was higher and that of other five components except for nodakenin was lower under pathological conditions. Additionally, the absorptive amount of nodakenin, osthole, 5-O-methylvisammioside and liquiritin was increased and that of ferulic acid and liquiritigenin was reduced in the RA rats than in the normal rats. Compared with the normal rats, the retention time of nodakenin, ferulic acid and liquiritin was reduced in vivo, whereas the retention time of osthole, 5-O-methylvisammioside and liquiritigenin was raised in the body for the RA rats. In contrast to the normal rats, the data demonstrated an increase in the elimination velocity of nodakenin and a decrease in the elimination velocity of the other five components except for nodakenin in the pathological state. Conclusion This study showed that the pharmacokinetic behavior of the six components, nodakenin, osthole, 5-O-methylvisammioside, ferulic acid, liquiritigenin, and liquiritin, is different in vivo between normal and pathological states of rats, and this research provided the necessary experimental data to explain the pharmacokinetics of QSD granules in both normal and pathological states and provide some references for its clinical application at some level.
Collapse
Affiliation(s)
- Xin Li
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Min Wang
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yuhong Zhong
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Qianqian Yin
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zheming Hu
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Wenli Tian
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhongyan Liu
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhidong Liu
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
3
|
Thomaz MDL, Vieira CP, Caris JA, Marques MP, Rocha A, Paz TA, Rezende REF, Lanchote VL. Liver Fibrosis Stages Affect Organic Cation Transporter 1/2 Activities in Hepatitis C Virus-Infected Patients. Pharmaceuticals (Basel) 2024; 17:865. [PMID: 39065716 PMCID: PMC11280093 DOI: 10.3390/ph17070865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 06/25/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
This study aims to evaluate the impact of liver fibrosis stages of chronic infection with hepatitis C virus (HCV) on the in vivo activity of organic cation transporters (hepatic OCT1 and renal OCT2) using metformin (MET) as a probe drug. Participants allocated in Group 1 (n = 15, mild to moderate liver fibrosis) or 2 (n = 13, advanced liver fibrosis and cirrhosis) received a single MET 50 mg oral dose before direct-acting antiviral (DAA) drug treatment (Phase 1) and 30 days after achieving sustained virologic response (Phase 2). OCT1/2 activity (MET AUC0-24) was found to be reduced by 25% when comparing the two groups in Phase 2 (ratio 0.75 (0.61-0.93), p < 0.05) but not in Phase 1 (ratio 0.81 (0.66-0.98), p > 0.05). When Phases 1 and 2 were compared, no changes were detected in both Groups 1 (ratio 1.10 (0.97-1.24), p > 0.05) and 2 (ratio 1.03 (0.94-1.12), p > 0.05). So, this study shows a reduction of approximately 25% in the in vivo activity of OCT1/2 in participants with advanced liver fibrosis and cirrhosis after achieving sustained virologic response and highlights that OCT1/2 in vivo activity depends on the liver fibrosis stage of chronic HCV infection.
Collapse
Affiliation(s)
- Matheus De Lucca Thomaz
- Department of Clinical Analysis, Food Science and Toxicology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil; (M.D.L.T.); (C.P.V.); (J.A.C.); (M.P.M.); (A.R.); (T.A.P.)
| | - Carolina Pinto Vieira
- Department of Clinical Analysis, Food Science and Toxicology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil; (M.D.L.T.); (C.P.V.); (J.A.C.); (M.P.M.); (A.R.); (T.A.P.)
| | - Juciene Aparecida Caris
- Department of Clinical Analysis, Food Science and Toxicology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil; (M.D.L.T.); (C.P.V.); (J.A.C.); (M.P.M.); (A.R.); (T.A.P.)
| | - Maria Paula Marques
- Department of Clinical Analysis, Food Science and Toxicology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil; (M.D.L.T.); (C.P.V.); (J.A.C.); (M.P.M.); (A.R.); (T.A.P.)
| | - Adriana Rocha
- Department of Clinical Analysis, Food Science and Toxicology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil; (M.D.L.T.); (C.P.V.); (J.A.C.); (M.P.M.); (A.R.); (T.A.P.)
| | - Tiago Antunes Paz
- Department of Clinical Analysis, Food Science and Toxicology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil; (M.D.L.T.); (C.P.V.); (J.A.C.); (M.P.M.); (A.R.); (T.A.P.)
| | - Rosamar Eulira Fontes Rezende
- Division of Gastroenterology, Department of Internal Medicine, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14049-900, Brazil;
- Reference Center, Hepatitis Outpatient Clinic, Municipal Health Secretary, Ribeirão Preto 14049-900, Brazil
| | - Vera Lucia Lanchote
- Department of Clinical Analysis, Food Science and Toxicology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil; (M.D.L.T.); (C.P.V.); (J.A.C.); (M.P.M.); (A.R.); (T.A.P.)
| |
Collapse
|
4
|
Chen Y, Ma F, Jones N, Deng R, Li C, Li C. Assessment of CYP3A-mediated drug interaction via cytokine (IL-6) elevation for mosunetuzumab using physiologically-based pharmacokinetic modeling. CPT Pharmacometrics Syst Pharmacol 2024; 13:234-246. [PMID: 38050329 PMCID: PMC10864933 DOI: 10.1002/psp4.13073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/06/2023] [Accepted: 10/19/2023] [Indexed: 12/06/2023] Open
Abstract
Mosunetuzumab is a CD3/CD20 bispecific antibody. As an on-target effect, transient elevation of interleukin-6 (IL-6) occurs in early treatment cycles. A physiologically-based pharmacokinetic (PBPK) model was developed to assess potential drug interaction caused by IL-6 enzyme suppression on cytochrome P450 3A (CYP3A) during mosunetuzumab treatment. The model's performance in predicting IL-6 CYP3A suppression and subsequent drug-drug interactions (DDIs) was verified using existing clinical data of DDIs caused by chronic and transient IL-6 elevation. Sensitivity analyses were performed for a complete DDI risk assessment. The IL-6 concentration- and time-dependent CYP3A suppression during mosunetuzumab treatment was simulated using PBPK model with incorporation of in vitro IL-6 inhibition data. At clinically approved doses/regimens, the DDI at maximum CYP3A suppression was predicted to be a midazolam maximum drug concentration in plasma (Cmax ) and area under the plasma drug concentration-time curve (AUC) ratio of 1.17 and 1.37, respectively. At the 95th percentile of IL-6 concentration level or when gut CYP3A suppression was considered, the predicted DDI risk for mosunetuzumab remained low (<2-fold). The PBPK-based DDI predictions informed the mosunetuzumab product label to monitor, in early cycles, the concentrations and toxicities for sensitive CYP3A substrates with narrow therapeutic windows.
Collapse
Affiliation(s)
- Yuan Chen
- Drug Metabolism and PharmacokineticsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Fang Ma
- Drug Metabolism and PharmacokineticsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Nicholas Jones
- Clinical ScienceGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Rong Deng
- Clinical PharmacologyGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Chunze Li
- Clinical PharmacologyGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Chi‐Chung Li
- Clinical PharmacologyGenentech, Inc.South San FranciscoCaliforniaUSA
| |
Collapse
|
5
|
Nakayama S, Toshimoto K, Yamazaki S, Snoeys J, Sugiyama Y. Physiologically-based pharmacokinetic modeling for investigating the effect of simeprevir on concomitant drugs and an endogenous biomarker of OATP1B. CPT Pharmacometrics Syst Pharmacol 2023; 12:1461-1472. [PMID: 37667529 PMCID: PMC10583237 DOI: 10.1002/psp4.13023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/11/2023] [Accepted: 07/18/2023] [Indexed: 09/06/2023] Open
Abstract
The orally available anti-hepatitis C virus (HCV) drug simeprevir exhibits nonlinear pharmacokinetics at the clinical doses due to saturation of cytochrome P450 (CYP) 3A4 metabolism and organic anion transporting peptide (OATP) 1B mediated hepatic uptake. Additionally, simeprevir increases exposures of concomitant drugs by CYP3A4 and OATP1B inhibition. The objective of this study was to develop physiologically-based pharmacokinetic (PBPK) models that could describe drug-drug interactions (DDIs) of simeprevir with concomitant drugs via CYP3A4 and OATP1B inhibition, and also to capture the effects on coproporphyrin-I (CP-I), an endogenous biomarker of OATP1B. PBPK modeling estimated unbound simeprevir inhibitory constant (Ki ) of 2.89 μM against CYP3A4 in the DDI results between simeprevir and midazolam in healthy volunteers. Then, we analyzed the DDIs between simeprevir and atorvastatin, a dual substrate of CYP3A4 and OATP1B, in healthy volunteers, and unbound Ki against OATP1B was estimated to be 0.00347 μM. Finally, we analyzed the increase in the blood level of CP-I by simeprevir to verify the Ki,OATP1B . Because CP-I was measured in subjects with HCV with various hepatic fibrosis state, Monte Carlo simulation was performed to involve the decreases in expression levels of hepatic CYP3A4 and OATP1B and their interindividual variabilities. The PBPK modeling coupled with Monte Carlo simulation using the Ki,OATP1B value obtained from atorvastatin study reasonably recovered the observed relationship between CP-I and simeprevir blood levels. In conclusion, the simeprevir PBPK model developed in this study can quantitatively describe the increase in exposures of concomitant drugs and an endogenous biomarker via inhibition of CYP3A4 and OATP1B.
Collapse
Affiliation(s)
- Shinji Nakayama
- DMPK Research Laboratories, Shoyaku, Innovative Research DivisionMitsubishi Tanabe Pharma CorporationYokohamaKanagawaJapan
| | - Kota Toshimoto
- Systems Pharmacology, Non‐Clinical Biomedical Science, Applied Research and OperationsAstellas Pharma Inc.IbarakiJapan
- Sugiyama Laboratory, RIKEN Cluster for ScienceRIKENYokohamaKanagawaJapan
| | - Shinji Yamazaki
- Drug Metabolism and PharmacokineticsJanssen Research and Development, LLCSan DiegoCaliforniaUSA
| | - Jan Snoeys
- Drug Metabolism and PharmacokineticsJanssen Research and DevelopmentBeerseBelgium
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Cluster for ScienceRIKENYokohamaKanagawaJapan
- Laboratory of Quantitative System Pharmacokinetics/PharmacodynamicsJosai International University (JIU)TokyoJapan
| |
Collapse
|
6
|
Abstract
The epidemic of obesity, type 2 diabetes and nonalcoholic liver disease (NAFLD) favors drug consumption, which augments the risk of adverse events including liver injury. For more than 30 years, a series of experimental and clinical investigations reported or suggested that the common pain reliever acetaminophen (APAP) could be more hepatotoxic in obesity and related metabolic diseases, at least after an overdose. Nonetheless, several investigations did not reproduce these data. This discrepancy might come from the extent of obesity and steatosis, accumulation of specific lipid species, mitochondrial dysfunction and diabetes-related parameters such as ketonemia and hyperglycemia. Among these factors, some of them seem pivotal for the induction of cytochrome P450 2E1 (CYP2E1), which favors the conversion of APAP to the toxic metabolite N-acetyl-p-benzoquinone imine (NAPQI). In contrast, other factors might explain why obesity and NAFLD are not always associated with more frequent or more severe APAP-induced acute hepatotoxicity, such as increased volume of distribution in the body, higher hepatic glucuronidation and reduced CYP3A4 activity. Accordingly, the occurrence and outcome of APAP-induced liver injury in an obese individual with NAFLD would depend on a delicate balance between metabolic factors that augment the generation of NAPQI and others that can mitigate hepatotoxicity.
Collapse
|
7
|
Wu W, Wu W, Ye Y, Li T, Wang B. mRNA and lncRNA expression profiles of liver tissues in children with biliary atresia. Exp Ther Med 2022; 24:634. [PMID: 36160912 PMCID: PMC9468840 DOI: 10.3892/etm.2022.11571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/01/2022] [Indexed: 02/05/2023] Open
Abstract
Progressive liver fibrosis is the most common phenotype in biliary atresia (BA). A number of pathways contribute to the fibrosis process so comprehensive understanding the mechanisms of liver fibrosis in BA will pave the way to improve patient's outcome after operation. In this study, the differentially expressed profiles of mRNAs and long non-coding RNAs from BA and choledochal cyst (CC) liver tissues were investigated and analyzed, which may provide potential clues to clarify hepatofibrosis mechanism in BA. A total of two BA and two CC liver tissue specimens were collected, the expression level of mRNAs and lncRNAs was detected by RNA sequencing. Differentially expressed mRNAs (DEmRNAs) were functionally annotated and protein-protein interaction networks (PPI) was established to predict the biological roles and interactive relationships. Differentially expressed lncRNAs (DElncRNAs) nearby targeted DEmRNA network and DElncRNA-DEmRNA co-expression network were constructed to further explore the roles of DElncRNAs in BA pathogenesis. The expression profiles of significant DEmRNAs were validated in Gene Expression Omnibus database. A total of 2,086 DEmRNAs and 184 DElncRNAs between BA and CC liver tissues were obtained. DEmRNAs were enriched in 521 Gene Ontology terms and 71 Kyoto Encyclopedia of Genes and Genomes terms which were mainly biological processes and metabolic pathways related to immune response and inflammatory response. A total of five hub proteins (TYRO protein tyrosine kinase binding protein, C-X-C motif chemokine ligand 8, pleckstrin, Toll-like receptor 8 and C-C motif chemokine receptor 5) were found in the PPI networks. A total of 31 DElncRNA-nearby-targeted DEmRNA pairs and 2,337 DElncRNA-DEmRNA co-expression pairs were obtained. The expression of DEmRNAs obtained from RNA sequencing were verified in GSE46960 dataset, generally. The present study identified key genes and lncRNAs participated in BA associated liver fibrosis, which may present a new avenue for understanding the patho-mechanism for hepatic fibrosis in BA.
Collapse
Affiliation(s)
- Wenyan Wu
- Medical Laboratory, Shenzhen Luohu People's Hospital, Shenzhen, Guangdong 518001, P.R. China
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523000, P.R. China
| | - Weifang Wu
- Medical College, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Department of General Surgery, Shenzhen Children's Hospital, Shenzhen, Guangdong 518026, P.R. China
| | - Yongqin Ye
- Department of General Surgery, Shenzhen Children's Hospital, Shenzhen, Guangdong 518026, P.R. China
- Faculty of Medicine, Macau University of Science and Technology, Macau SAR 999078, P.R. China
| | - Tao Li
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523000, P.R. China
| | - Bin Wang
- Department of General Surgery, Shenzhen Children's Hospital, Shenzhen, Guangdong 518026, P.R. China
- Correspondence to: Professor Bin Wang, Department of General Surgery, Shenzhen Children's Hospital, 7019 Yitian Road, Futian, Shenzhen, Guangdong 518026, P.R. China
| |
Collapse
|
8
|
Nies AT, Schaeffeler E, Schwab M. Hepatic solute carrier transporters and drug therapy: Regulation of expression and impact of genetic variation. Pharmacol Ther 2022; 238:108268. [DOI: 10.1016/j.pharmthera.2022.108268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/25/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022]
|
9
|
Lu Y, Wang Y, He Y, Pan J, Jin Y, Zheng L, Huang Y, Li Y, Liu W. Aidi injection altered the activity of CYP2D4, CYP1A2, CYP2C19, CYP3A2, CYP2E1 and CYP2C11 in normal and diethylnitrosamine-induced hepatocellular carcinoma in rats. JOURNAL OF ETHNOPHARMACOLOGY 2022; 286:114930. [PMID: 34952190 DOI: 10.1016/j.jep.2021.114930] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 12/16/2021] [Accepted: 12/16/2021] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Aidi injection (ADI), a traditional chinese medicine preparation, is widely used in combination with chemotherapy for the treatment of various malignant tumors, such as hepatocellular carcinoma (HCC). Studies have shown that changes in cytochrome P450 (CYP450) activity in disease states would affect the metabolism of drugs in vivo, especially liver diseases. However, the changes of Aidi injection on the activities of CYP2D4, CYP1A2, CYP2C19, CYP3A2, CYP2E1 and CYP2C11 in normal and HCC states are still unknown. AIM OF THE STUDY The cocktail probe drugs method was used to investigate the effects of ADI on the activity of CYP2D4, CYP1A2, CYP2C19, CYP3A2, CYP2E1 and CYP2C11 in normal and HCC rats. MATERIALS AND METHODS The HCC rats was induced by diethylnitrosamine (DEN). Then, both normal and HCC rats were randomly divided into 2 groups (n = 6). They were given saline or ADI (10 mL/kg/d, i.p) for 2 weeks, respectively. On the fifteenth day, cocktail probe mixing solution, including metoprolol (10 mg/kg), caffeine (1.0 mg/kg), omeprazole (2.0 mg/kg), midazolam (2.0 mg/kg), chlorzoxazone (4.0 mg/kg) and tolbutamide (0.5 mg/kg), was injected into tail vein of all rats in each group. The blood sample was obtained at specified time. After the protein is precipitated, six probe drugs are analyzed by ultra performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS). RESULTS Compared with control group, the activity of CYP3A2 and CYP2E1 was significantly lower in the ADI group. Compared with the model group, the activities of CYP1A2, CYP3A2, CYP2E1, and CYP2C11 enzymes in the ADI model group were significantly reduced. Additionally, the activity of CYP2D4, CYP1A2, CYP2C19, CYP3A2, CYP2E1 and CYP2C11 enzymes in model group was significantly lower than control group. CONCLUSIONS ADI can inhibit a lot of CYP450 enzyme, so it may reduce the dosage of chemotherapeutic drugs to reach the required plasma concentration of chemotherapeutic drugs, which is of great significance for the combination of anti-tumor chemotherapeutic drugs and is worthy of further in-depth study and clinical attention.
Collapse
Affiliation(s)
- Yuan Lu
- The Affiliated Hospital of Guizhou Medical University, 28(#) Guiyi Road, Guiyang, 550004, Guizhou, China; State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, 550004, China; School of Pharmacy, Guizhou Medical University, No.9, Beijing Road, Yunyan District, Guiyang, 550004, China
| | - Yanli Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, 550004, China; School of Pharmacy, Guizhou Medical University, No.9, Beijing Road, Yunyan District, Guiyang, 550004, China
| | - Yan He
- The Affiliated Hospital of Guizhou Medical University, 28(#) Guiyi Road, Guiyang, 550004, Guizhou, China
| | - Jie Pan
- Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, 550004, China
| | - Yang Jin
- School of Pharmacy, Guizhou Medical University, No.9, Beijing Road, Yunyan District, Guiyang, 550004, China
| | - Lin Zheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, 550004, China
| | - Yong Huang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, 550004, China
| | - Yongjun Li
- Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, 550004, China; School of Pharmacy, Guizhou Medical University, No.9, Beijing Road, Yunyan District, Guiyang, 550004, China
| | - Wen Liu
- The Affiliated Hospital of Guizhou Medical University, 28(#) Guiyi Road, Guiyang, 550004, Guizhou, China; School of Pharmacy, Guizhou Medical University, No.9, Beijing Road, Yunyan District, Guiyang, 550004, China.
| |
Collapse
|
10
|
Shapses M, Tang L, Layne A, Beri A, Rotman Y. Fatty Liver Is an Independent Risk Factor for Delayed Recovery from Anesthesia. Hepatol Commun 2021; 5:1848-1859. [PMID: 34558821 PMCID: PMC8557314 DOI: 10.1002/hep4.1772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/31/2021] [Accepted: 06/09/2021] [Indexed: 11/17/2022] Open
Abstract
Fatty liver (FL) is associated with altered activity of hepatic drug-metabolizing enzymes, but the clinical significance is unknown. Many anesthetic agents are metabolized in the liver. We aimed to determine whether FL impacts recovery from anesthesia as a surrogate for altered drug metabolism. This was a single-center, retrospective, case-control study of all adults who underwent anesthesia and concurrent abdominal imaging (n = 2,021) in a hospital setting. FL (n = 234) was identified through radiology reports. Anesthesia recovery, the primary endpoint, was defined by Aldrete's recovery score (RS, 0-10), assessed following postanesthesia care unit (PACU) arrival, with RS ≥8 considered discharge eligible. FL and controls were compared using univariate and multivariate analyses, adjusting for confounders. A secondary matched-pairs analysis matched FL and controls 1:1 for confounders. Time from airway removal to discharge eligibility was compared using multivariate Cox regression. On PACU arrival, 54.1% of FL were discharge eligible compared to 61.7% of controls (P = 0.03), with lower activity scores on univariate (P = 0.03) and multivariate analysis (P = 0.03). On matched-pairs analysis, discharge eligibility, activity, consciousness, and total RSs were lower in FL (P ≤ 0.04 for all). Median time from airway removal to discharge eligibility was 43% longer in FL (univariate, P = 0.01; multivariate hazard ratio, 1.32; P = 0.046). To further exclude confounding by obesity, we performed a sensitivity analysis limited to a body mass index <30, where FL was still associated with lower activity (P = 0.03) and total RS (P = 0.03). Conclusion: Patients with FL have delayed recovery from anesthesia, suggesting altered drug metabolism independent of metabolic risk factors.
Collapse
Affiliation(s)
- Mark Shapses
- Liver and Energy Metabolism SectionLiver Diseases BranchNational Institute of Diabetes and Digestive and Kidney DiseasesNational Institutes of HealthBethesdaMDUSA
| | - Lin Tang
- Department of Perioperative MedicineNational Institutes of Health Clinical CenterBethesdaMDUSA
| | - Austin Layne
- Biomedical Translational Research Information SystemNational Institutes of Health Clinical CenterBethesdaMDUSA
| | - Andrea Beri
- Biomedical Translational Research Information SystemNational Institutes of Health Clinical CenterBethesdaMDUSA
| | - Yaron Rotman
- Liver and Energy Metabolism SectionLiver Diseases BranchNational Institute of Diabetes and Digestive and Kidney DiseasesNational Institutes of HealthBethesdaMDUSA
| |
Collapse
|
11
|
Tu Y, Wang L, Rong Y, Tam V, Yin T, Gao S, Singh R, Hu M. Hepatoenteric recycling is a new disposition mechanism for orally administered phenolic drugs and phytochemicals in rats. eLife 2021; 10:e58820. [PMID: 34196607 PMCID: PMC8248983 DOI: 10.7554/elife.58820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 06/16/2021] [Indexed: 12/28/2022] Open
Abstract
Many orally administered phenolic drugs undergo enterohepatic recycling (EHR), presumably mediated by the hepatic phase II enzymes. However, the disposition of extrahepatically generated phase II metabolites is unclear. This paper aims to determine the new roles of liver and intestine in the disposition of oral phenolics. Sixteen representative phenolics were tested using direct portal vein infusion and/or intestinal perfusion. The results showed that certain glucuronides were efficiently recycled by liver. OATP1B1/1B3/2B1 were the responsible uptake transporters. Hepatic uptake is the rate-limiting step in hepatic recycling. Our findings showed that the disposition of many oral phenolics is mediated by intestinal glucuronidation and hepatic recycling. A new disposition mechanism 'Hepatoenteric Recycling (HER)", where intestine is the metabolic organ and liver is the recycling organ, was revealed. Further investigations focusing on HER should help interpret how intestinal aliments or co-administered drugs that alter gut enzymes (e.g. UGTs) expression/activities will impact the disposition of phenolics.
Collapse
Affiliation(s)
- Yifan Tu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of HoustonHoustonUnited States
| | - Lu Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of HoustonHoustonUnited States
| | - Yi Rong
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of HoustonHoustonUnited States
| | - Vincent Tam
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of HoustonHoustonUnited States
| | - Taijun Yin
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of HoustonHoustonUnited States
| | - Song Gao
- Department of Pharmaceutical Sciences, College of Pharmacy, Texas Southern UniversityHoustonUnited States
| | - Rashim Singh
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of HoustonHoustonUnited States
| | - Ming Hu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of HoustonHoustonUnited States
| |
Collapse
|
12
|
Pippa LF, Vieira CP, Caris JA, Rocha A, Garcia CP, Rezende REF, Lanchote VL. Clinical treatment for hepatitis C reverses CYP2C19 inhibition. Br J Clin Pharmacol 2021; 87:4013-4019. [PMID: 33738827 DOI: 10.1111/bcp.14829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 12/15/2022] Open
Abstract
AIMS Infection by the hepatitis C virus (HCV) generates inflammatory response selectively modulating cytochrome P450 protein (CYP) activities. This study assessed the effect of chronic hepatitis C on CYP2C19 activity in patients with HCV. METHODS Patients with HCV infection (n = 23) at different fibrosis stages were allocated into groups 1 (F0/F1 and F2, mild to moderate fibrosis) and 2 (F3 and F4, advanced fibrosis stages). Phase 1 was conducted before the treatment with direct-acting antivirals (DAAs) and phase 2 after the sustained virological response. Participants were administered 2 mg of a single oral dose of omeprazole (OME) as probe drug in both phases. Metabolic ratios (MRs) (plasma samples collected at 4 h after OME administration) were calculated by dividing plasma concentrations of 5-hydroxyomeprazole by OME. RESULTS The MRs for group 1 were 0.45 (0.34-0.60, 90% confidence interval) and 0.69 (0.50-0.96) for phases 1 and 2, respectively, while the MRs for group 2 were 0.25 (0.21-0.31) and 0.41 (0.30-0.56) for phases 1 and 2, respectively. MRs were different (P < .05) between phases 1 and 2 for both groups, as well as between groups 1 and 2 in phase 1, but not in phase 2 (P > .05). CONCLUSIONS Both groups presented different MRs before and after treatment with DAAs, evidencing that CYP2C19 inhibition during inflammation was at least partially reversed after DAA treatment. Groups 1 and 2 were also found to be different in phase 1 but not phase 2, showing that CYP2C19 metabolic activity does not differ between groups after DAA treatment.
Collapse
Affiliation(s)
- Leandro Francisco Pippa
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Carolina Pinto Vieira
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Juciene Aparecida Caris
- Department of Neurosciences and Behavioural Sciences, School of Medicine of Ribeirão Preto, University of São Paulo, Brazil
| | - Adriana Rocha
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Camile Prates Garcia
- Department of Internal Medicine, Division of Gastroenterology, School of Medicine of Ribeirão Preto, University of São Paulo, Brazil
| | - Rosamar Eulira Fontes Rezende
- Department of Internal Medicine, Division of Gastroenterology, School of Medicine of Ribeirão Preto, University of São Paulo, Brazil.,Reference Centre, Hepatitis Outpatient Clinic, Municipal Health Secretary, Ribeirão Preto, São Paulo, Brazil
| | - Vera Lucia Lanchote
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
13
|
Drug-Drug Interactions Involving Intestinal and Hepatic CYP1A Enzymes. Pharmaceutics 2020; 12:pharmaceutics12121201. [PMID: 33322313 PMCID: PMC7764576 DOI: 10.3390/pharmaceutics12121201] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/02/2020] [Accepted: 12/08/2020] [Indexed: 12/29/2022] Open
Abstract
Cytochrome P450 (CYP) 1A enzymes are considerably expressed in the human intestine and liver and involved in the biotransformation of about 10% of marketed drugs. Despite this doubtless clinical relevance, CYP1A1 and CYP1A2 are still somewhat underestimated in terms of unwanted side effects and drug–drug interactions of their respective substrates. In contrast to this, many frequently prescribed drugs that are subjected to extensive CYP1A-mediated metabolism show a narrow therapeutic index and serious adverse drug reactions. Consequently, those drugs are vulnerable to any kind of inhibition or induction in the expression and function of CYP1A. However, available in vitro data are not necessarily predictive for the occurrence of clinically relevant drug–drug interactions. Thus, this review aims to provide an up-to-date summary on the expression, regulation, function, and drug–drug interactions of CYP1A enzymes in humans.
Collapse
|
14
|
Ozawa S, Miura T, Terashima J, Habano W, Ishida S. Recent Progress in Prediction Systems for Drug-induced Liver Injury Using in vitro Cell Culture. Drug Metab Lett 2020; 14:25-40. [PMID: 33267768 DOI: 10.2174/1872312814666201202112610] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/26/2020] [Accepted: 11/03/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND In order to avoid drug-induced liver injury (DILI), in vitro assays, which enable the assessment of both metabolic activation and immune reaction processes that ultimately result in DILI, are needed. OBJECTIVE In this study, the recent progress in the application of in vitro assays using cell culture systems is reviewed for potential DILI-causing drugs/xenobiotics and a mechanistic study on DILI, as well as for the limitations of in vitro cell culture systems for DILI research. METHODS Information related to DILI was collected through a literature search of the PubMed database. RESULTS The initial biological event for the onset of DILI is the formation of cellular protein adducts after drugs have been metabolically activated by drug metabolizing enzymes. The damaged peptides derived from protein adducts lead to the activation of CD4+ helper T lymphocytes and recognition by CD8+ cytotoxic T lymphocytes, which destroy hepatocytes through immunological reactions. Because DILI is a major cause of drug attrition and drug withdrawal, numerous in vitro systems consisting of hepatocytes and immune/inflammatory cells, or spheroids of human primary hepatocytes containing non-parenchymal cells have been developed. These cellular-based systems have identified DILIinducing drugs with approximately 50% sensitivity and 90% specificity. CONCLUSION Different co-culture systems consisting of human hepatocyte-derived cells and other immune/inflammatory cells have enabled the identification of DILI-causing drugs and of the actual mechanisms of action.
Collapse
Affiliation(s)
- Shogo Ozawa
- Department of Clinical Pharmaceutical Sciences, Division of Pharmacodynamics and Molecular Genetics, School of Pharmacy, Iwate Medical University, Yahaba. Japan
| | - Toshitaka Miura
- Department of Clinical Pharmaceutical Sciences, Division of Pharmacodynamics and Molecular Genetics, School of Pharmacy, Iwate Medical University, Yahaba. Japan
| | - Jun Terashima
- Department of Clinical Pharmaceutical Sciences, Division of Pharmacodynamics and Molecular Genetics, School of Pharmacy, Iwate Medical University, Yahaba. Japan
| | - Wataru Habano
- Department of Clinical Pharmaceutical Sciences, Division of Pharmacodynamics and Molecular Genetics, School of Pharmacy, Iwate Medical University, Yahaba. Japan
| | - Seiichi Ishida
- Department of Pharmacology, National Institute of Health Sciences, Kawasaki. Japan
| |
Collapse
|
15
|
D'souza S, Lau KCK, Coffin CS, Patel TR. Molecular mechanisms of viral hepatitis induced hepatocellular carcinoma. World J Gastroenterol 2020; 26:5759-5783. [PMID: 33132633 PMCID: PMC7579760 DOI: 10.3748/wjg.v26.i38.5759] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/03/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023] Open
Abstract
Chronic infection with viral hepatitis affects half a billion individuals worldwide and can lead to cirrhosis, cancer, and liver failure. Liver cancer is the third leading cause of cancer-associated mortality, of which hepatocellular carcinoma (HCC) represents 90% of all primary liver cancers. Solid tumors like HCC are complex and have heterogeneous tumor genomic profiles contributing to complexity in diagnosis and management. Chronic infection with hepatitis B virus (HBV), hepatitis delta virus (HDV), and hepatitis C virus (HCV) are the greatest etiological risk factors for HCC. Due to the significant role of chronic viral infection in HCC development, it is important to investigate direct (viral associated) and indirect (immune-associated) mechanisms involved in the pathogenesis of HCC. Common mechanisms used by HBV, HCV, and HDV that drive hepatocarcinogenesis include persistent liver inflammation with an impaired antiviral immune response, immune and viral protein-mediated oxidative stress, and deregulation of cellular signaling pathways by viral proteins. DNA integration to promote genome instability is a feature of HBV infection, and metabolic reprogramming leading to steatosis is driven by HCV infection. The current review aims to provide a brief overview of HBV, HCV and HDV molecular biology, and highlight specific viral-associated oncogenic mechanisms and common molecular pathways deregulated in HCC, and current as well as emerging treatments for HCC.
Collapse
Affiliation(s)
- Simmone D'souza
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary T2N 1N4, AB, Canada
| | - Keith CK Lau
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary T2N 1N4, AB, Canada
| | - Carla S Coffin
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary T2N 1N4, AB, Canada
| | - Trushar R Patel
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary T2N 1N4, AB, Canada
- Department of Chemistry and Biochemistry, Alberta RNA Research and Training Institute, University of Lethbridge, Lethbridge T1K3M4, AB, Canada
| |
Collapse
|
16
|
Suzuki Y, Sasamoto Y, Koyama T, Yoshijima C, Nakatochi M, Kubo M, Momozawa Y, Uehara R, Ohno K. Substantially Increased Plasma Coproporphyrin-I Concentrations Associated With OATP1B1*15 Allele in Japanese General Population. Clin Transl Sci 2020; 14:382-388. [PMID: 32961019 PMCID: PMC7877856 DOI: 10.1111/cts.12889] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 08/19/2020] [Indexed: 12/13/2022] Open
Abstract
Coproporphyrin-I (CP-I) in plasma is a sensitive and specific endogenous probe for phenotyping organic anion transporting polypeptides 1B (OATP1B, encoded by SLCO1B). A few small-scale studies suggested that plasma CP-I concentration is affected by OATP1B1 polymorphism, but detailed studies are lacking. In this large-scale study, we measured plasma CP-I concentrations in 391 subjects from the Japanese general population, and evaluated the relationship between plasma CP-I concentrations and OATP1B1 polymorphisms to further assess the utility of plasma CP-I concentrations as an endogenous OATP1B probe. Plasma CP-I concentrations were 0.45 ± 0.12, 0.47 ± 0.16, 0.47 ± 0.20, 0.50 ± 0.15, 0.54 ± 0.14, and 0.74 ± 0.31 ng/mL in participants with OATP1B1*1b/*1b (n = 103), *1a/*1b (n = 122), *1a/*1a (n = 40), *1b/*15 (n = 74), *1a/*15 (n = 41), and *15/*15 (n = 11), respectively, showing an ascending rank order with significant difference (P < 0.0001). Post hoc analysis revealed significant increases in plasma CP-I concentration in OATP1B1*1b/*15 (P = 0.036), *1a/*15 (P = 0.0005), and *15/*15 (P = 0.0003) groups compared with the OATP1B1*1b/*1b group. There was no significant difference among OATP1B genotypes in plasma concentration of 3-carboxy-4-methyl-5-propyl-2-furanpropanoic acid, a uremic toxin reported to decrease OATP1B activity in vivo. These findings confirm the utility of plasma CP-I concentrations as an endogenous biomarker for phenotyping of OATP1B activity. Plasma CP-I concentration is potentially useful for the study of drug-drug interactions via OATP1B or individual dose adjustment of OATP1B substrates.
Collapse
Affiliation(s)
- Yosuke Suzuki
- Department of Medication Use Analysis and Clinical Research, Meiji Pharmaceutical University, Tokyo, Japan
| | - Yuri Sasamoto
- Department of Medication Use Analysis and Clinical Research, Meiji Pharmaceutical University, Tokyo, Japan
| | - Teruhide Koyama
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Chisato Yoshijima
- Department of Medication Use Analysis and Clinical Research, Meiji Pharmaceutical University, Tokyo, Japan
| | - Masahiro Nakatochi
- Department of Nursing, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Michiaki Kubo
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yukihide Momozawa
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Ritei Uehara
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Keiko Ohno
- Department of Medication Use Analysis and Clinical Research, Meiji Pharmaceutical University, Tokyo, Japan
| |
Collapse
|
17
|
Rodrigues AD, Rowland A. Profiling of Drug-Metabolizing Enzymes and Transporters in Human Tissue Biopsy Samples: A Review of the Literature. J Pharmacol Exp Ther 2020; 372:308-319. [PMID: 31879375 DOI: 10.1124/jpet.119.262972] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 12/19/2019] [Indexed: 03/08/2025] Open
Abstract
Within the drug pharmacokinetics (PK)-absorption, distribution, metabolism, and excretion (ADME) research community, investigators regularly generate in vitro data sets using appropriately vendor-sourced and processed human tissue. Such data enable drug screening, the generation of kinetic parameters, extrapolation of in vitro to in vivo, as well as the modeling and simulation of drug PK. Although there are large numbers of manuscripts describing studies with deceased organ donor tissue, relatively few investigators have published studies utilizing living donor tissue biopsy samples. After a review of the available literature, it was possible to find publications describing the use of tissue biopsy samples to determine enzyme inhibition ex vivo, the study of genotype-phenotype associations, the evaluation of tissue expression profiling following an inducer, and assessment of correlations between tissue expression profiles and in vivo-derived trait measures (e.g., biomarker plasma levels and probe drug PK). Some reports described multiple single-tissue biopsies, whereas others described single multiple-organ biopsies. It is concluded that biopsy-derived data can support modeling exercises (as input data and when validating models) and enable the assessment of organ-specific changes in enzyme and transporter profiles resulting from drug interactions, disease (e.g., metabolic disease, fibrosis, inflammation, cancer, infection), age, pregnancy, organ impairment, and genotype. With the emergence of multiorgan axes (e.g., microbiome-gut-liver-kidney) and interest in remote sensing (interorgan communication), it is envisioned that there will be increased demand for single- and multiorgan tissue biopsy data to support hypothesis testing and PK-ADME model building. SIGNIFICANCE STATEMENT: Based on a review of the literature, it is apparent that profiling of human tissue biopsy samples is useful in support of pharmacokinetics (PK)-absorption, distribution, metabolism, and excretion (ADME)-related studies. With conventional tissue biopsy as precedent, it is envisioned that researchers will turn to less invasive "liquid biopsy" methods in support of ADME-related studies (e.g., profiling of plasma-derived tissue-specific nanovesicles). Generation of such multiorgan liquid biopsy data in larger numbers of subjects and at multiple study time points will provide a rich data set for modeling purposes.
Collapse
Affiliation(s)
- A David Rodrigues
- ADME Sciences, Medicine Design, Worldwide Research & Development, Pfizer Inc., Groton, Connecticut (D.R.) and College of Medicine and Public Health, Flinders University, Adelaide, Australia (A.R.)
| | - Andrew Rowland
- ADME Sciences, Medicine Design, Worldwide Research & Development, Pfizer Inc., Groton, Connecticut (D.R.) and College of Medicine and Public Health, Flinders University, Adelaide, Australia (A.R.)
| |
Collapse
|
18
|
Sevastianos VA, Voulgaris TA, Dourakis SP. Hepatitis C, systemic inflammation and oxidative stress: correlations with metabolic diseases. Expert Rev Gastroenterol Hepatol 2020; 14:27-37. [PMID: 31868062 DOI: 10.1080/17474124.2020.1708191] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Hepatitis C chronic infection has long been correlated with numerous systemic diseases, such as diabetes mellitus and hepatic steatosis. Recent studies have also revealed an association with atherosclerosis.Areas covered: An analysis is presented on the mechanisms through which the hepatitis C viral infection can lead to a systemic increase in pro-inflammatory markers, especially tumor necrosis factor-a and interleukin-6. The immunological imbalance created may, through different mechanisms, act on the metabolic pathways that contribute to the development of insulin resistance, the accumulation of lipids in the liver, and even the formation of atherosclerotic plaques. Moreover, an additional contributing factor to the above-mentioned metabolic derangements is the unopposed oxidative stress observed in chronic hepatitis C viral infection. The virus itself contributes to the formation of oxidative stress, through alterations in the trace metal homeostasis and its effect on pro-inflammatory cytokines, such as tumor necrosis factor-a.Expert opinion: The scope of this review is to emphasize the importance of the metabolic manifestations of hepatitis C viral infection and to elucidate the pathophysiological mechanisms behind their emergence.
Collapse
Affiliation(s)
- Vassilios A Sevastianos
- Department of Internal Medicine and Liver Outpatient Clinic, "Evangelismos" General Hospital, Athens, Greece
| | - Theodoros A Voulgaris
- Department of Internal Medicine and Liver Outpatient Clinic, "Evangelismos" General Hospital, Athens, Greece
| | - Spyros P Dourakis
- Department of Internal Μedicine, Medical School, National and Kapodistrian University of Athens, General Hospital of Athens Ippokrateio, Athens, Greece
| |
Collapse
|
19
|
Yan Y, Allweiss L, Yang D, Kang J, Wang J, Qian X, Zhang T, Liu H, Wang L, Liu S, Sui J, Chen X, Dandri M, Zhao J, Lu F. Down-regulation of cell membrane localized NTCP expression in proliferating hepatocytes prevents hepatitis B virus infection. Emerg Microbes Infect 2019; 8:879-894. [PMID: 31179847 PMCID: PMC6567113 DOI: 10.1080/22221751.2019.1625728] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Hepatocyte proliferation could result in the loss of covalently closed circular DNA (cccDNA) and the emergence of cccDNA-cleared nascent hepatocytes, which appear refractory to hepatitis B virus (HBV) reinfection with unknown mechanism(s). Sodium taurocholate cotransporting polypeptide (NTCP) is the functional receptor for HBV entry. In this study, down-regulation of cell membrane localized NTCP expression in proliferating hepatocytes was found to prevent HBV infection in HepG2-NTCP-tet cells and in liver-humanized mice. In patients, lower NTCP protein expression was correlated well with higher levels of hepatocyte proliferation and less HBsAg expression in HBV-related focal nodular hyperplasia (FNH) tissues. Clinically, significantly lower NTCP protein expression was correlated with more active hepatocyte proliferation in CHB patients with severe active necroinflammation and better antiviral treatment outcome. Mechanistically, the activation of cell cycle regulatory genes p53, S-phase kinase-associated protein 2 (SKP2) and cyclin D1 during cell proliferation, as well as proliferative and inflammatory cytokine Interleukin-6 (IL-6) could transcriptionally down-regulate NTCP expression. From these aspects, we conclude that within the milieu of hepatocyte proliferation, down-regulation of cell membrane localized NTCP expression level renders nascent hepatocytes resistant to HBV reinfection. This may accelerate virus clearance during immune-mediated cell death and compensatory proliferation of survival hepatocytes.
Collapse
Affiliation(s)
- Ying Yan
- a State Key Laboratory of Natural and Biomimetic Drugs, Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences , Peking University Health Science Center , Beijing , People's Republic of China
| | - Lena Allweiss
- b Department of Medicine, Center for Internal Medicine , University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Danli Yang
- a State Key Laboratory of Natural and Biomimetic Drugs, Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences , Peking University Health Science Center , Beijing , People's Republic of China
| | - Jingting Kang
- c Institute of Basic Medical Sciences Chinese Academy of Medical Sciences , School of Basic Medicine Peking Union Medical College , Beijing , People's Republic of China
| | - Jianwen Wang
- a State Key Laboratory of Natural and Biomimetic Drugs, Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences , Peking University Health Science Center , Beijing , People's Republic of China
| | - Xiangjun Qian
- a State Key Laboratory of Natural and Biomimetic Drugs, Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences , Peking University Health Science Center , Beijing , People's Republic of China
| | - Ting Zhang
- a State Key Laboratory of Natural and Biomimetic Drugs, Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences , Peking University Health Science Center , Beijing , People's Republic of China
| | - Hui Liu
- a State Key Laboratory of Natural and Biomimetic Drugs, Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences , Peking University Health Science Center , Beijing , People's Republic of China
| | - Lu Wang
- a State Key Laboratory of Natural and Biomimetic Drugs, Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences , Peking University Health Science Center , Beijing , People's Republic of China
| | - Shuhong Liu
- d Department of Pathology and Hepatology , The 5th Medical Centre, Chinese PLA General Hospital , Beijing , People's Republic of China
| | - Jianhua Sui
- e Biologics Research Center , National Institute of Biological Sciences , Beijing , People's Republic of China
| | - Xiangmei Chen
- a State Key Laboratory of Natural and Biomimetic Drugs, Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences , Peking University Health Science Center , Beijing , People's Republic of China
| | - Maura Dandri
- b Department of Medicine, Center for Internal Medicine , University Medical Center Hamburg-Eppendorf , Hamburg , Germany.,f German Center for Infection Research (DZIF) , Hamburg-Lübeck-Borstel-Riems Partner Site , Hamburg , Germany
| | - Jingmin Zhao
- d Department of Pathology and Hepatology , The 5th Medical Centre, Chinese PLA General Hospital , Beijing , People's Republic of China
| | - Fengmin Lu
- a State Key Laboratory of Natural and Biomimetic Drugs, Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences , Peking University Health Science Center , Beijing , People's Republic of China
| |
Collapse
|
20
|
Kurzawski M, Szeląg-Pieniek S, Łapczuk-Romańska J, Wrzesiński M, Sieńko J, Oswald S, Droździk M. The reference liver - ABC and SLC drug transporters in healthy donor and metastatic livers. Pharmacol Rep 2019; 71:738-745. [PMID: 31207436 DOI: 10.1016/j.pharep.2019.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 03/29/2019] [Accepted: 04/08/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Analysis of results and conclusions in studies dedicated to pathology of the liver are usually based on comparison of pathological liver specimens and control/reference (considered as healthy) tissues. There are two main sources of the control liver samples used as the reference livers, i.e. deceased organ donor livers and non-tumorous tissue from metastatic livers, which are also applied for drug transporter investigations. However, no information has yet been published on drug transporters in these two major types of reference livers. METHODS We explored ABC (P-gp, MRP1, MRP2, MRP3, MRP4, BCRP, BSEP) and SLC (NTCP, MCT1, OCT1, OCT3, OAT2, OATP1B1, OATP1B3, OATP2B1) family transporters expression (qPCR) and protein abundance (LC-MS/MS) in healthy donors (n = 9) and metastatic (n = 13) livers. RESULTS The analysis of mRNA content revealed significant differences in ABCB11, ABCC1, ABCG2, SLC10A1, SLC16A1, SLCO1B1 and SLCO2B1 gene expression between livers from organ donors and patients who underwent surgical resection of metastatic tumors. The protein abundance of NTCP was significantly higher, whereas of P-gp significantly lower in non-tumorous tissues from metastatic livers. Greater inter-individual variability in protein abundance of all studied transporters in subjects with metastatic colon cancer was also observed. CONCLUSIONS The results suggest that final conclusions in liver pathology studies may depend on the reference liver tissue used, especially in gene expression studies.
Collapse
Affiliation(s)
- Mateusz Kurzawski
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Szczecin, Poland.
| | - Sylwia Szeląg-Pieniek
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Szczecin, Poland
| | - Joanna Łapczuk-Romańska
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Szczecin, Poland
| | - Maciej Wrzesiński
- Department of General and Transplantation Surgery, Marie Curie Regional Hospital, Szczecin, Poland
| | - Jerzy Sieńko
- Department of General and Transplantation Surgery, Pomeranian Medical University, Szczecin, Poland
| | - Stefan Oswald
- Department of Clinical Pharmacology, University Medicine of Greifswald, Greifswald, Germany
| | - Marek Droździk
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
21
|
Development and Application of an UHPLC-MS/MS Method for Comparative Pharmacokinetic Study of Eight Major Bioactive Components from Yin Chen Hao Tang in Normal and Acute Liver Injured Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:3239785. [PMID: 30519262 PMCID: PMC6241247 DOI: 10.1155/2018/3239785] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 10/10/2018] [Indexed: 12/12/2022]
Abstract
Yin Chen Hao Tang (YCHT) is one of the most famous hepatoprotective herbal formulas in China, but its pharmacokinetic investigation in model rats has been rarely conducted. In this study, the hepatic injury model was caused by intraperitoneal injections of carbon tetrachloride (CCl4), and YCHT was orally administered to the model and normal rats. An ultrahigh performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) method was established to analyze the plasma pharmacokinetics of eight major bioactive ingredients from YCHT in both the normal and liver injured rats. The calibration curves presented good linearity (r > 0.9981) in the concentration range. The relative standard deviation (RSD%) of inter- and intraday precision was within 9.55%, and the accuracy (RE%) ranged from -10.72% to 2.46%. The extraction recovery, matrix effect, and stability were demonstrated to be within acceptable ranges. The lower limit of detection (LLOD) and lower limit of quantitation (LLOQ) were around 0.1 ng/mL and 0.5 ng/mL, respectively, which were much lower than those in other related researches. Results reveal that there are significant differences in the pharmacokinetics of scoparone, geniposide, rhein, aloe-emodin, physcion, and chrysophanol in hepatic injured rats as compared to those in control except for scopoletin and emodin. Our experimental results provide a meaningful reference for the clinical dosage of YCHT in treating liver disorders, and the improvement of LLOD and LLOQ can also broaden the range of our method's application, which is very suitable for quantitating these eight compounds with low levels.
Collapse
|
22
|
Howard M, Barber J, Alizai N, Rostami-Hodjegan A. Dose adjustment in orphan disease populations: the quest to fulfill the requirements of physiologically based pharmacokinetics. Expert Opin Drug Metab Toxicol 2018; 14:1315-1330. [PMID: 30465453 DOI: 10.1080/17425255.2018.1546288] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION While the media is engaged and fascinated by the idea of 'Precision Medicine', the nuances related to 'Precision Dosing' seem to be largely ignored. Assuming the 'right drug' is selected, clinicians still need to decide on the 'right dose' for individuals. Ideally, optimal dosing should be studied in clinical trials; however, many drugs on the market lack evidence-based dosing recommendations, and small groups of patients (orphan disease populations) are dependent on local guidance and clinician experience to determine drug dosage adjustments. Areas Covered: This report explores the current understanding of dosing adjustment in special populations and examines the requirements for developing 'in silico' models for pediatric, elderly and pregnant patients. The report also highlights current use of modeling to provide evidence-based recommendations for drug labeling in the absence of complete clinical trials in orphan disease populations. Expert Opinion: Physiologically based pharmacokinetics (PBPK) is an attractive prospect for determining the best drug dosage adjustments in special populations. However, it is not sufficient for individualized, or even stratified dosing, unless the systems (drug-independent) data required to build robust PBPK models are obtained. Such models are not a substitute for clinical trials, but they are an alternative to undocumented and inconsistent guesswork.
Collapse
Affiliation(s)
- Martyn Howard
- a Centre for Applied Pharmacokinetic Research , University of Manchester , Manchester , UK
| | - Jill Barber
- a Centre for Applied Pharmacokinetic Research , University of Manchester , Manchester , UK
| | - Naved Alizai
- b Leeds General Infirmary , Leeds Children's Hospital , Leeds , UK
| | - Amin Rostami-Hodjegan
- a Centre for Applied Pharmacokinetic Research , University of Manchester , Manchester , UK
| |
Collapse
|
23
|
Uchida Y, Naiki K, Kouyama JI, Sugawara K, Nakao M, Motoya D, Inao M, Nakayama N, Imai Y, Tomiya T, Mochida S. Serum asunaprevir concentrations showing correlation with the extent of liver fibrosis as a factor inducing liver injuries in patients with genotype-1b hepatitis C virus receiving daclatasvir plus asunaprevir therapy. PLoS One 2018; 13:e0205600. [PMID: 30308053 PMCID: PMC6181393 DOI: 10.1371/journal.pone.0205600] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 09/27/2018] [Indexed: 12/14/2022] Open
Abstract
AIMS Liver injury can occur during antiviral therapies with direct-acting antivirals (DAAs), potentially necessitating discontinuation of the therapies, with consequent worsening of the sustained viral response (SVR) rates, in patients with hepatitis C virus (HCV). To clarify the mechanisms involved in serum transaminase level elevation, we performed a retrospective evaluation of the serum concentrations of daclatasvir and asunaprevir, both classified as DAAs, in patients receiving treatment with a combination of the two drugs. METHODS Subjects were 278 Japanese patients with genotype-1b HCV who received daclatasvir plus asunaprevir therapy for more than 4 weeks. Serum concentrations of both the DAAs were measured at 4 weeks after the initiation of therapy. RESULT Liver injuries including serum AST and/or ALT level elevation to 150 U/L or over were found in 34 patients (12.2%). Multivariate logistic regression analysis identified serum asunaprevir concentrations as being significantly associated with developing liver injury, with an odds ratio of 1.046 (95% confidence interval 1.011-1.082, p<0.05). Serum asunaprevir concentrations showed correlation with the extent of liver fibrosis, estimated by peripheral platelets counts and serum albumin levels and baseline and FIB4 index and serum Mac-2 binding protein glycosylation isomer (M2BPGi) levels at 4 weeks of the therapy; the concentrations were significantly higher among patients showing 3.0 or more of M2BPGi levels than among those with the levels less than 3.0; on the other hand, no such correlation/difference was found in serum daclatasvir concentrations. CONCLUSION High serum concentrations of serum asunaprevir, which were associated with the extent of liver fibrosis, appear to provoke the occurrence of liver injury in patients with genotype-1b HCV receiving combined daclatasvir plus asunaprevir therapy.
Collapse
Affiliation(s)
- Yoshihito Uchida
- Department of Gastroenterology & Hepatology, Faculty of Medicine, Saitama Medical University, Moroyama, Saitama, Japan
| | - Kayoko Naiki
- Department of Gastroenterology & Hepatology, Faculty of Medicine, Saitama Medical University, Moroyama, Saitama, Japan
| | - Jun-ichi Kouyama
- Department of Gastroenterology & Hepatology, Faculty of Medicine, Saitama Medical University, Moroyama, Saitama, Japan
| | - Kayoko Sugawara
- Department of Gastroenterology & Hepatology, Faculty of Medicine, Saitama Medical University, Moroyama, Saitama, Japan
| | - Masamitsu Nakao
- Department of Gastroenterology & Hepatology, Faculty of Medicine, Saitama Medical University, Moroyama, Saitama, Japan
| | - Daisuke Motoya
- Department of Gastroenterology & Hepatology, Faculty of Medicine, Saitama Medical University, Moroyama, Saitama, Japan
| | - Mie Inao
- Department of Gastroenterology & Hepatology, Faculty of Medicine, Saitama Medical University, Moroyama, Saitama, Japan
| | - Nobuaki Nakayama
- Department of Gastroenterology & Hepatology, Faculty of Medicine, Saitama Medical University, Moroyama, Saitama, Japan
| | - Yukinori Imai
- Department of Gastroenterology & Hepatology, Faculty of Medicine, Saitama Medical University, Moroyama, Saitama, Japan
| | - Tomoaki Tomiya
- Department of Gastroenterology & Hepatology, Faculty of Medicine, Saitama Medical University, Moroyama, Saitama, Japan
| | - Satoshi Mochida
- Department of Gastroenterology & Hepatology, Faculty of Medicine, Saitama Medical University, Moroyama, Saitama, Japan
| |
Collapse
|
24
|
Fattahi S, Karimi Alivije M, Babamahmoodi F, Bayani M, Sadeghi Haddad Zavareh M, Asouri M, Lotfi M, Amirbozorgi G, Akhavan-Niaki H. Cytochrome P450 Genes (CYP2E1 and CYP1A1) Variants and Susceptibility to Chronic Hepatitis B Virus Infection. Indian J Clin Biochem 2018; 33:467-472. [PMID: 30319195 PMCID: PMC6170240 DOI: 10.1007/s12291-017-0698-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 09/11/2017] [Indexed: 01/26/2023]
Abstract
Hepatitis B virus (HBV) infection is a worldwide health concern which is associated with significant morbidity and mortality. Both viral and host factors have a significant effect on infection, replication and pathogenesis of HBV. The aim of this study was to investigate the effect of CYP2E1 and CYP1A1 genetic variants on susceptibility to HBV. 143 individuals including 54 chronic HBV patients and 89 healthy controls were enrolled in the genotyping procedure. rs2031920 and rs3813867 at CYP2E1 as well as rs4646421 and rs2198843 at CYP1A1 loci were studied in all subjects using PCR-RFLP (restriction fragment length polymorphism) analysis. Both variants at CYP2E1 locus were monomorphic in all studied subjects. Genotype frequency of rs4646421 was significantly different between chronic HBV patients and healthy blood donors (P = 0.04, OR 4.31; 95% CI 1.04-17.7). Furthermore, individuals carrying at least one C allele (CC or CT genotypes) for rs4646421 seemed to have a decrease risk of hepatitis in comparison with TT genotype (P = 0.039). Our results showed a relationship between rs4646421 TT genotype (rare genotype) and the risk for developing chronic HBV infection (four times higher). Further studies are needed to examine the role of CYP1A1 polymorphism in susceptibility to chronic HBV infection.
Collapse
Affiliation(s)
- Sadegh Fattahi
- Cellular and Molecular Department, North Research Center, Pasteur Institute of Iran, Amol, Iran
| | | | - Farhang Babamahmoodi
- Department of Antimicrobial Resistance Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Masomeh Bayani
- Infectious Diseases and Tropical Medicine Research Center, Babol University of Medical Sciences, Babol, Iran
| | | | - Mohsen Asouri
- Cellular and Molecular Department, North Research Center, Pasteur Institute of Iran, Amol, Iran
| | - Maryam Lotfi
- Cellular and Molecular Department, North Research Center, Pasteur Institute of Iran, Amol, Iran
| | - Galia Amirbozorgi
- Cellular and Molecular Department, North Research Center, Pasteur Institute of Iran, Amol, Iran
| | - Haleh Akhavan-Niaki
- Cellular and Molecular Department, North Research Center, Pasteur Institute of Iran, Amol, Iran
- Department of Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
25
|
Sauer M, Haubner C, Richter G, Ehler J, Mencke T, Mitzner S, Margraf S, Altrichter J, Doß S, Nöldge-Schomburg G. Impaired Cell Viability and Functionality of Hepatocytes After Incubation With Septic Plasma-Results of a Second Prospective Biosensor Study. Front Immunol 2018; 9:1448. [PMID: 29988573 PMCID: PMC6026797 DOI: 10.3389/fimmu.2018.01448] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 06/11/2018] [Indexed: 12/14/2022] Open
Abstract
Liver dysfunction (LD) and liver failure are associated with poor outcome in critically ill patients. In patients with severe sepsis or septic shock, LD occurred in nearly 19% of patients. An early diagnosis of LD at time of initial damage of the liver can lead to a better prognosis of these patients because an early start of therapy is possible. We performed a second prospective study with septic patients to test a new cell-based cytotoxicity device (biosensor) to evaluate clinical relevance for early diagnosis of LD and prognostic capacity. In the clinical study, 99 intensive care unit patients were included in two groups. From the patients of the septic group (n = 51, SG), and the control (non-septic) group [n = 49, control group (CG)] were drawn 20 ml blood at inclusion, after 3, and 7 days for testing with the biosensor. Patients’ data were recorded for hospital survival, organ function, and demographic data, illness severity [acute physiology and chronic health evaluation (APACHE) II-, sepsis-related organ failure assessment (SOFA) scores], cytokines, circulating-free deoxyribonucleic acid/neutrophil-derived extracellular traps (cf-DNA/NETs), microbiological results, and pre-morbidity. For the developed cytotoxicity test, the human liver cell line HepG2/C3A was used. Patients’ plasma was incubated in a microtiter plate assay with the test cells and after 6 days incubation the viability (trypan blue staining, XTT-test) and functionality (synthesis of albumin, cytochrome 1A2 activity) was analyzed. An impairment of viability and functionality of test cells was only seen in the SG compared with the CG. The plasma of non-survivors in the SG led to a more pronounced impairment of test cells than the plasma of survivors at inclusion. In addition, the levels of cf-DNA/NETs were significantly higher in the SG at inclusion, after 3, and after 7 days compared with the CG. The SG showed an in-hospital mortality of 24% and the values of bilirubin, APACHE II-, and SOFA scores were markedly higher at inclusion than in the CG. Hepatotoxicity of septic plasma was already detected with the liver cell-based biosensor at inclusion and also in the course of disease. The biosensor may be a tool for early diagnosis of LD in septic patients and may have prognostic relevance.
Collapse
Affiliation(s)
- Martin Sauer
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Rostock, Rostock, Germany.,Extracorporeal Immunomodulation (EXIM), Fraunhofer Institute for Cell Therapy and Immunology, Rostock, Germany
| | - Cristof Haubner
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Rostock, Rostock, Germany
| | - Georg Richter
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Rostock, Rostock, Germany
| | - Johannes Ehler
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Rostock, Rostock, Germany
| | - Thomas Mencke
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Rostock, Rostock, Germany
| | - Steffen Mitzner
- Extracorporeal Immunomodulation (EXIM), Fraunhofer Institute for Cell Therapy and Immunology, Rostock, Germany.,Division of Nephrology, Department of Medicine, University Hospital of Rostock, Rostock, Germany
| | - Stefan Margraf
- Extracorporeal Immunomodulation (EXIM), Fraunhofer Institute for Cell Therapy and Immunology, Rostock, Germany
| | - Jens Altrichter
- Division of Nephrology, Department of Medicine, University Hospital of Rostock, Rostock, Germany
| | - Sandra Doß
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Rostock, Rostock, Germany
| | - Gabriele Nöldge-Schomburg
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Rostock, Rostock, Germany
| |
Collapse
|
26
|
Okubo H, Ando H, Sorin Y, Nakadera E, Fukada H, Morishige J, Miyazaki A, Ikejima K. Gadoxetic acid-enhanced magnetic resonance imaging to predict paritaprevir-induced hyperbilirubinemia during treatment of hepatitis C. PLoS One 2018; 13:e0196747. [PMID: 29709031 PMCID: PMC5927452 DOI: 10.1371/journal.pone.0196747] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 03/01/2018] [Indexed: 02/06/2023] Open
Abstract
Background Paritaprevir inhibits organic anion–transporting polypeptide (OATP)1B1 and OATP1B3, which transport bilirubin. Hyperbilirubinemia is an adverse event reported during hepatitis C treatment. Gadoxetic acid is also transported by OATP1B1/1B3. We evaluated whether the enhancement effect in gadoxetic acid–enhanced magnetic resonance (MR) imaging could predict the plasma concentration of paritaprevir and might anticipate the development of hyperbilirubinemia. Methods This prospective study evaluated 27 patients with hepatitis C who underwent gadoxetic acid–enhanced MR imaging prior to treatment with ombitasvir, paritaprevir, and ritonavir. The contrast enhancement index (CEI), a measure of liver enhancement during the hepatobiliary phase, was assessed. Plasma trough concentrations, and concentrations at 2, 4, and 6 h after dosing were determined 7 d after the start of treatment. Results Seven patients (26%) developed hyperbilirubinemia (≥ 1.6 mg/dl). Paritaprevir trough concentration (Ctrough) was significantly higher in patients with hyperbilirubinemia than in those without (p = 0.022). We found an inverse relationship between CEI and Ctrough (r = 0.612, p = 0.001), while there was not a significantly weak inverse relationship between AUC0–6 h and CEI (r = −0.338, p = 0.085). The partial correlation coefficient between CEI and Ctrough was −0.425 (p = 0.034), while excluding the effects of albumin and the FIB-4 index. Receiver operating characteristic (ROC) curve analysis showed that the CEI was relatively accurate in predicting hyperbilirubinemia, with area under the ROC of 0.882. Multivariate analysis showed that the CEI < 1.61 was the only independent predictor related to the development of hyperbilirubinemia, with an odds ratio of 9.08 (95% confidence interval 1.05–78.86, p = 0.046). Conclusions Hepatic enhancement with gadoxetic acid was independently related to paritaprevir concentration and was an independent pretreatment factor in predicting hyperbilirubinemia. Gadoxetic acid–enhanced MR imaging can therefore be useful in determining the risk of paritaprevir-induced hyperbilirubinemia.
Collapse
Affiliation(s)
- Hironao Okubo
- Department of Gastroenterology, Juntendo University Nerima Hospital, Tokyo, Japan
- * E-mail:
| | - Hitoshi Ando
- Department of Cellular and Molecular Function Analysis, Kanazawa University Graduate School of Medical Sciences, Ishikawa, Japan
| | - Yushi Sorin
- Department of Gastroenterology, Juntendo University Nerima Hospital, Tokyo, Japan
| | - Eisuke Nakadera
- Department of Gastroenterology, Juntendo University Nerima Hospital, Tokyo, Japan
| | - Hiroo Fukada
- Department of Gastroenterology, Juntendo University Nerima Hospital, Tokyo, Japan
| | - Junichi Morishige
- Department of Cellular and Molecular Function Analysis, Kanazawa University Graduate School of Medical Sciences, Ishikawa, Japan
| | - Akihisa Miyazaki
- Department of Gastroenterology, Juntendo University Nerima Hospital, Tokyo, Japan
| | - Kenichi Ikejima
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
27
|
Elucidating the Plasma and Liver Pharmacokinetics of Simeprevir in Special Populations Using Physiologically Based Pharmacokinetic Modelling. Clin Pharmacokinet 2018; 56:781-792. [PMID: 27896690 DOI: 10.1007/s40262-016-0476-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The disposition of simeprevir (SMV) in humans is characterised by cytochrome P450 3A4 metabolism and hepatic uptake by organic anion transporting polypeptide 1B1/3 (OATP1B1/3). This study was designed to investigate SMV plasma and liver exposure upon oral administration in subjects infected with hepatitis C virus (HCV), in subjects of Japanese or Chinese origin, subjects with organ impairment and subjects with OATP genetic polymorphisms, using physiologically based pharmacokinetic modelling. Simulations showed that compared with healthy Caucasian subjects, SMV plasma exposure was 2.4-, 1.7-, 2.2- and 2.0-fold higher, respectively, in HCV-infected Caucasian subjects, in healthy Japanese, healthy Chinese and subjects with severe renal impairment. Further simulations showed that compared with HCV-infected Caucasian subjects, SMV plasma exposure was 1.6-fold higher in HCV-infected Japanese subjects. In subjects with OATP1B1 genetic polymorphisms, no noteworthy changes in SMV pharmacokinetics were observed. Simulations suggested that liver concentrations in Caucasians with HCV are 18 times higher than plasma concentrations.
Collapse
|
28
|
Coutant DE, Hall SD. Disease-Drug Interactions in Inflammatory States via Effects on CYP-Mediated Drug Clearance. J Clin Pharmacol 2018; 58:849-863. [DOI: 10.1002/jcph.1093] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 01/11/2018] [Indexed: 12/17/2022]
Affiliation(s)
- David E. Coutant
- Department of Drug Disposition; Eli Lilly and Company; Indianapolis IN USA
| | - Stephen D. Hall
- Department of Drug Disposition; Eli Lilly and Company; Indianapolis IN USA
| |
Collapse
|
29
|
Billington S, Ray AS, Salphati L, Xiao G, Chu X, Humphreys WG, Liao M, Lee CA, Mathias A, Hop CECA, Rowbottom C, Evers R, Lai Y, Kelly EJ, Prasad B, Unadkat JD. Transporter Expression in Noncancerous and Cancerous Liver Tissue from Donors with Hepatocellular Carcinoma and Chronic Hepatitis C Infection Quantified by LC-MS/MS Proteomics. Drug Metab Dispos 2018; 46:189-196. [PMID: 29138286 PMCID: PMC5776333 DOI: 10.1124/dmd.117.077289] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 10/19/2017] [Indexed: 12/11/2022] Open
Abstract
Protein expression of major hepatobiliary drug transporters (NTCP, OATPs, OCT1, BSEP, BCRP, MATE1, MRPs, and P-gp) in cancerous (C, n = 8) and adjacent noncancerous (NC, n = 33) liver tissues obtained from patients with chronic hepatitis C with hepatocellular carcinoma (HCV-HCC) were quantified by LC-MS/MS proteomics. Herein, we compare our results with our previous data from noninfected, noncirrhotic (control, n = 36) and HCV-cirrhotic (n = 30) livers. The amount of membrane protein yielded from NC and C HCV-HCC tissues decreased (31%, 67%) relative to control livers. In comparison with control livers, with the exception of NTCP, MRP2, and MATE1, transporter expression decreased in NC (38%-76%) and C (56%-96%) HCV-HCC tissues. In NC HCV-HCC tissues, NTCP expression increased (113%), MATE1 expression decreased (58%), and MRP2 expression was unchanged relative to control livers. In C HCV-HCC tissues, NTCP and MRP2 expression decreased (63%, 56%) and MATE1 expression was unchanged relative to control livers. Compared with HCV-cirrhotic livers, aside from NTCP, OCT1, BSEP, and MRP2, transporter expression decreased in NC (41%-71%) and C (54%-89%) HCV-HCC tissues. In NC HCV-HCC tissues, NTCP and MRP2 expression increased (362%, 142%), whereas OCT1 and BSEP expression was unchanged. In C HCV-HCC tissues, OCT1 and BSEP expression decreased (90%, 80%) relative to HCV-cirrhotic livers, whereas NTCP and MRP2 expression was unchanged. Expression of OATP2B1, BSEP, MRP2, and MRP3 decreased (56%-72%) in C HCV-HCC tissues in comparison with matched NC tissues (n = 8), but the expression of other transporters was unchanged. These data will be helpful in the future to predict transporter-mediated hepatocellular drug concentrations in patients with HCV-HCC.
Collapse
Affiliation(s)
- Sarah Billington
- Department of Pharmaceutics, University of Washington, Seattle, Washington (S.B., E.J.K., B.P., J.D.U.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Inc., Foster City, California (A.S.R., A.M., Y.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (G.X., C.R.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey (X.C., R.E.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and Translational Sciences, Ardea Biosciences, Inc., San Diego, California (C.A.L.)
| | - Adrian S Ray
- Department of Pharmaceutics, University of Washington, Seattle, Washington (S.B., E.J.K., B.P., J.D.U.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Inc., Foster City, California (A.S.R., A.M., Y.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (G.X., C.R.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey (X.C., R.E.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and Translational Sciences, Ardea Biosciences, Inc., San Diego, California (C.A.L.)
| | - Laurent Salphati
- Department of Pharmaceutics, University of Washington, Seattle, Washington (S.B., E.J.K., B.P., J.D.U.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Inc., Foster City, California (A.S.R., A.M., Y.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (G.X., C.R.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey (X.C., R.E.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and Translational Sciences, Ardea Biosciences, Inc., San Diego, California (C.A.L.)
| | - Guangqing Xiao
- Department of Pharmaceutics, University of Washington, Seattle, Washington (S.B., E.J.K., B.P., J.D.U.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Inc., Foster City, California (A.S.R., A.M., Y.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (G.X., C.R.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey (X.C., R.E.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and Translational Sciences, Ardea Biosciences, Inc., San Diego, California (C.A.L.)
| | - Xiaoyan Chu
- Department of Pharmaceutics, University of Washington, Seattle, Washington (S.B., E.J.K., B.P., J.D.U.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Inc., Foster City, California (A.S.R., A.M., Y.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (G.X., C.R.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey (X.C., R.E.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and Translational Sciences, Ardea Biosciences, Inc., San Diego, California (C.A.L.)
| | - W Griffith Humphreys
- Department of Pharmaceutics, University of Washington, Seattle, Washington (S.B., E.J.K., B.P., J.D.U.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Inc., Foster City, California (A.S.R., A.M., Y.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (G.X., C.R.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey (X.C., R.E.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and Translational Sciences, Ardea Biosciences, Inc., San Diego, California (C.A.L.)
| | - Mingxiang Liao
- Department of Pharmaceutics, University of Washington, Seattle, Washington (S.B., E.J.K., B.P., J.D.U.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Inc., Foster City, California (A.S.R., A.M., Y.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (G.X., C.R.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey (X.C., R.E.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and Translational Sciences, Ardea Biosciences, Inc., San Diego, California (C.A.L.)
| | - Caroline A Lee
- Department of Pharmaceutics, University of Washington, Seattle, Washington (S.B., E.J.K., B.P., J.D.U.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Inc., Foster City, California (A.S.R., A.M., Y.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (G.X., C.R.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey (X.C., R.E.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and Translational Sciences, Ardea Biosciences, Inc., San Diego, California (C.A.L.)
| | - Anita Mathias
- Department of Pharmaceutics, University of Washington, Seattle, Washington (S.B., E.J.K., B.P., J.D.U.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Inc., Foster City, California (A.S.R., A.M., Y.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (G.X., C.R.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey (X.C., R.E.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and Translational Sciences, Ardea Biosciences, Inc., San Diego, California (C.A.L.)
| | - Cornelis E C A Hop
- Department of Pharmaceutics, University of Washington, Seattle, Washington (S.B., E.J.K., B.P., J.D.U.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Inc., Foster City, California (A.S.R., A.M., Y.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (G.X., C.R.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey (X.C., R.E.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and Translational Sciences, Ardea Biosciences, Inc., San Diego, California (C.A.L.)
| | - Christopher Rowbottom
- Department of Pharmaceutics, University of Washington, Seattle, Washington (S.B., E.J.K., B.P., J.D.U.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Inc., Foster City, California (A.S.R., A.M., Y.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (G.X., C.R.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey (X.C., R.E.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and Translational Sciences, Ardea Biosciences, Inc., San Diego, California (C.A.L.)
| | - Raymond Evers
- Department of Pharmaceutics, University of Washington, Seattle, Washington (S.B., E.J.K., B.P., J.D.U.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Inc., Foster City, California (A.S.R., A.M., Y.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (G.X., C.R.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey (X.C., R.E.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and Translational Sciences, Ardea Biosciences, Inc., San Diego, California (C.A.L.)
| | - Yurong Lai
- Department of Pharmaceutics, University of Washington, Seattle, Washington (S.B., E.J.K., B.P., J.D.U.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Inc., Foster City, California (A.S.R., A.M., Y.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (G.X., C.R.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey (X.C., R.E.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and Translational Sciences, Ardea Biosciences, Inc., San Diego, California (C.A.L.)
| | - Edward J Kelly
- Department of Pharmaceutics, University of Washington, Seattle, Washington (S.B., E.J.K., B.P., J.D.U.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Inc., Foster City, California (A.S.R., A.M., Y.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (G.X., C.R.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey (X.C., R.E.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and Translational Sciences, Ardea Biosciences, Inc., San Diego, California (C.A.L.)
| | - Bhagwat Prasad
- Department of Pharmaceutics, University of Washington, Seattle, Washington (S.B., E.J.K., B.P., J.D.U.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Inc., Foster City, California (A.S.R., A.M., Y.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (G.X., C.R.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey (X.C., R.E.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and Translational Sciences, Ardea Biosciences, Inc., San Diego, California (C.A.L.)
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington (S.B., E.J.K., B.P., J.D.U.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Inc., Foster City, California (A.S.R., A.M., Y.L.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (G.X., C.R.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey (X.C., R.E.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); and Translational Sciences, Ardea Biosciences, Inc., San Diego, California (C.A.L.)
| |
Collapse
|
30
|
Atilano-Roque A, Roda G, Fogueri U, Kiser JJ, Joy MS. Effect of Disease Pathologies on Transporter Expression and Function. J Clin Pharmacol 2017; 56 Suppl 7:S205-21. [PMID: 27385176 DOI: 10.1002/jcph.768] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 05/08/2016] [Accepted: 05/10/2016] [Indexed: 12/12/2022]
Abstract
Transporters are important determinants of drug absorption, distribution, and excretion. The clinical relevance of drug transporters in drug disposition and toxicology depends on their localization in liver, kidney, and brain. There has been growing evidence regarding the importance of disease status on alterations in metabolizing enzymes and transporter proteins. This review focuses on uptake and efflux transporter proteins in liver, kidney, and brain and discusses mechanisms of altered transporter expression and function secondary to disease.
Collapse
Affiliation(s)
- Amandla Atilano-Roque
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA
| | - Gavriel Roda
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA
| | - Uma Fogueri
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA
| | - Jennifer J Kiser
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA
| | - Melanie S Joy
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA.,Division of Renal Diseases and Hypertension, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
31
|
Sheng RF, Wang HQ, Yang L, Jin KP, Xie YH, Fu CX, Zeng MS. Assessment of liver fibrosis using T1 mapping on Gd-EOB-DTPA-enhanced magnetic resonance. Dig Liver Dis 2017; 49:789-795. [PMID: 28237298 DOI: 10.1016/j.dld.2017.02.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 02/02/2017] [Accepted: 02/06/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Few studies have investigated the value of Gd-EOB-DTPA-enhanced T1 mapping in exact fibrosis staging, especially its correlation with hepatic molecular transporters. AIMS To investigate the diagnostic value of Gd-EOB-DTPA-enhanced T1 mapping in staging liver fibrosis and its relationship with hepatic molecular transporters. METHODS Thirty rats were divided into the carbon tetrachloride-induced fibrosis groups and a control group. T1-mapping was performed before and 20min after administration of Gd-EOB-DTPA. The T1 relaxation time and reduction rate (Δ%) were calculated, and their correlations with the degree of fibrosis, necroinflammatory activity, iron load and hepatic molecular transporters were assessed and compared. RESULTS Hepatobiliary phase T1 relaxation time (HBP) and Δ% were different between each adjacent fibrosis subgroups(P=0.000-0.042). Very strong correlations existed between fibrosis and both HBP and Δ% (r=0.960/-0.952), and multivariate analyses revealed that fibrosis was the only factor independently predicted by HBP (P=0.000) and Δ% (P=0.001), comparing to necroinflammatory activity and iron load. The expression of the organic anion transporting polypeptide1a1 (Oatp1a1) was significantly correlated with HBP and Δ% at both mRNA (r=-0.741/0.697) and protein (r=-0.577/0.602) levels. Weaker correlations were found for multidrug resistance associated protein2 (Mrp2). Generally, both transporters showed decreasing levels with increasing degrees of fibrosis. CONCLUSION Gd-EOB-DTPA-enhanced T1 mapping may provide a reliable diagnostic tool in staging liver fibrosis, and can be regarded as a useful imaging biomarker of hepatocyte transporter function.
Collapse
Affiliation(s)
- Ruo Fan Sheng
- Department of Radiology, Zhongshan Hospital, Fudan University, Xuhui District, Shanghai, China; Shanghai Institute of Medical Imaging, Xuhui District, Shanghai, China
| | - He Qing Wang
- Department of Radiology, Zhongshan Hospital, Fudan University, Xuhui District, Shanghai, China; Shanghai Institute of Medical Imaging, Xuhui District, Shanghai, China
| | - Li Yang
- Department of Radiology, Zhongshan Hospital, Fudan University, Xuhui District, Shanghai, China; Shanghai Institute of Medical Imaging, Xuhui District, Shanghai, China
| | - Kai Pu Jin
- Department of Radiology, Zhongshan Hospital, Fudan University, Xuhui District, Shanghai, China; Shanghai Institute of Medical Imaging, Xuhui District, Shanghai, China
| | - Yan Hong Xie
- Department of Pathology, Zhongshan Hospital, Fudan University, Xuhui District, Shanghai 200032, China
| | - Cai Xia Fu
- MR Collaboration NEA, Siemens Ltd. China, Shanghai, China
| | - Meng Su Zeng
- Department of Radiology, Zhongshan Hospital, Fudan University, Xuhui District, Shanghai, China; Shanghai Institute of Medical Imaging, Xuhui District, Shanghai, China.
| |
Collapse
|
32
|
王 鹤, 孙 鹏, 刘 克. 肝脏转运体表达和功能的变化对肝疾病的影响. Shijie Huaren Xiaohua Zazhi 2017; 25:1427-1437. [DOI: 10.11569/wcjd.v25.i16.1427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
转运体是药物吸收、分布、代谢和排泄的重要决定因素, 在肝脏表达尤为广泛. 肝脏转运体可以摄取大多数内源性物质、营养物质和外源性物质进入肝脏, 在肝脏内经过一系列的代谢转化, 最终将其外排入胆汁, 并由胆汁排到肝外. 越来越多的证据表明, 肝脏疾病状态下转运体的表达和功能会发生改变, 影响药物在体内的处置过程, 进而增加药物相互作用的可能性, 同时加大了疾病药物治疗的难度. 本文从肝脏摄取型和外排型转运体两方面出发, 针对肝脏转运体表达和功能的变化对肝疾病的影响作一综述.
Collapse
|
33
|
Effect of Steady-State Faldaprevir on Pharmacokinetics of Atorvastatin or Rosuvastatin in Healthy Volunteers: A Prospective Open-Label, Fixed-Sequence Crossover Study. J Clin Pharmacol 2017; 57:1305-1314. [DOI: 10.1002/jcph.931] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 04/03/2017] [Indexed: 12/12/2022]
|
34
|
Thakkar N, Slizgi JR, Brouwer KLR. Effect of Liver Disease on Hepatic Transporter Expression and Function. J Pharm Sci 2017; 106:2282-2294. [PMID: 28465155 DOI: 10.1016/j.xphs.2017.04.053] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 04/20/2017] [Accepted: 04/21/2017] [Indexed: 12/27/2022]
Abstract
Liver disease can alter the disposition of xenobiotics and endogenous substances. Regulatory agencies such as the Food and Drug Administration and the European Medicines Evaluation Agency recommend, if possible, studying the effect of liver disease on drugs under development to guide specific dose recommendations in these patients. Although extensive research has been conducted to characterize the effect of liver disease on drug-metabolizing enzymes, emerging data have implicated that the expression and function of hepatobiliary transport proteins also are altered in liver disease. This review summarizes recent developments in the field, which may have implications for understanding altered disposition, safety, and efficacy of new and existing drugs. A brief review of liver physiology and hepatic transporter localization/function is provided. Then, the expression and function of hepatic transporters in cholestasis, hepatitis C infection, hepatocellular carcinoma, human immunodeficiency virus infection, nonalcoholic fatty liver disease and nonalcoholic steatohepatitis, and primary biliary cirrhosis are reviewed. In the absence of clinical data, nonclinical information in animal models is presented. This review aims to advance the understanding of altered expression and function of hepatic transporters in liver disease and the implications of such changes on drug disposition.
Collapse
Affiliation(s)
- Nilay Thakkar
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Jason R Slizgi
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Kim L R Brouwer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599.
| |
Collapse
|
35
|
Dietrich CG, Rau M, Jahn D, Geier A. Changes in drug transport and metabolism and their clinical implications in non-alcoholic fatty liver disease. Expert Opin Drug Metab Toxicol 2017; 13:625-640. [PMID: 28359183 DOI: 10.1080/17425255.2017.1314461] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The incidence of non-alcoholic fatty liver disease (NAFLD) is rising, especially in Western countries. Drug treatment in patients with NAFLD is common since it is linked to other conditions like diabetes, obesity, and cardiovascular disease. Consequently, changes in drug metabolism may have serious clinical implications. Areas covered: A literature search for studies in animal models or patients with obesity, fatty liver, non-alcoholic steatohepatitis (NASH) or NASH cirrhosis published before November 2016 was performed. After discussing epidemiology and animal models for NAFLD, we summarized both basic as well as clinical studies investigating changes in drug transport and metabolism in NAFLD. Important drug groups were assessed separately with emphasis on clinical implications for drug treatment in patients with NAFLD. Expert opinion: Given the frequency of NAFLD even today, a high degree of drug treatment in NAFLD patients appears safe and well-tolerated despite considerable changes in hepatic uptake, distribution, metabolism and transport of drugs in these patients. NASH causes changes in biliary excretion, systemic concentrations, and renal handling of drugs leading to alterations in drug efficacy or toxicity under specific circumstances. Future clinical drug studies should focus on this special patient population in order to avoid serious adverse events in NAFLD patients.
Collapse
Affiliation(s)
- Christoph G Dietrich
- a Bethlehem Center of Health , Department of Medicine , Stolberg/Rhineland , Germany
| | - Monika Rau
- b Division of Hepatology, Department of Medicine II , University of Würzburg , Würzburg , Germany
| | - Daniel Jahn
- b Division of Hepatology, Department of Medicine II , University of Würzburg , Würzburg , Germany
| | - Andreas Geier
- b Division of Hepatology, Department of Medicine II , University of Würzburg , Würzburg , Germany
| |
Collapse
|
36
|
Lawitz E, Poordad F, Gutierrez JA, Kakuda TN, Picchio G, Beets G, Vandevoorde A, Van Remoortere P, Jacquemyn B, Luo D, Ouwerkerk-Mahadevan S, Vijgen L, Van Eygen V, Beumont M. Simeprevir, daclatasvir and sofosbuvir for hepatitis C virus-infected patients with decompensated liver disease. J Viral Hepat 2017; 24:287-294. [PMID: 27878906 DOI: 10.1111/jvh.12645] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 10/04/2016] [Indexed: 01/08/2023]
Abstract
Approximately three million individuals in the United States are chronically infected with hepatitis C virus (HCV). Chronic HCV infection may lead to the development of compensated as well as decompensated liver cirrhosis. The Phase II IMPACT study was conducted in HCV genotype 1- or 4-infected cirrhotic patients with portal hypertension or decompensated liver disease and assessed for the first time the combination of the three direct-acting antivirals simeprevir, daclatasvir and sofosbuvir. Treatment-naïve or treatment-experienced adults with Child-Pugh (CP) score <7 (CP A) and evidence of portal hypertension, or CP score 7-9 (CP B), received 12 weeks of simeprevir 150 mg, daclatasvir 60 mg and sofosbuvir 400 mg, once daily. The primary efficacy endpoint was sustained virologic response 12 weeks after end of treatment (SVR12). Pharmacokinetics and safety were also assessed. Overall, 40 patients were enrolled (CP A: 19; CP B: 21). All 40 patients achieved SVR12. At week 8, the mean pharmacokinetic exposure to simeprevir, sofosbuvir, daclatasvir and GS-331007 (sofosbuvir metabolite) was 2.2-, 1.5-, 1.2- and 1.2-fold higher in patients with CP B than CP A, respectively. Grade 1/2 adverse events (AEs) occurred in 26 of 40 (65%) patients. One CP B patient had a Grade 3 AE (gastrointestinal haemorrhage), which was reported as a serious AE but not considered related to study drugs. Treatment for 12 weeks with simeprevir, daclatasvir and sofosbuvir was generally safe and well tolerated, and resulted in 100% of cirrhotic patients with portal hypertension or decompensated liver disease achieving SVR12.
Collapse
Affiliation(s)
- E Lawitz
- Texas Liver Institute, University of Texas Health Science Center, San Antonio, TX, USA
| | - F Poordad
- Texas Liver Institute, University of Texas Health Science Center, San Antonio, TX, USA
| | - J A Gutierrez
- Texas Liver Institute, University of Texas Health Science Center, San Antonio, TX, USA
| | | | - G Picchio
- Janssen Research & Development LLC, Raritan, NJ, USA
| | - G Beets
- Janssen Pharmaceutica NV, Beerse, Belgium
| | | | | | | | - D Luo
- Janssen Research & Development LLC, Titusville, NJ, USA
| | | | - L Vijgen
- Janssen Pharmaceutica NV, Beerse, Belgium
| | | | - M Beumont
- Janssen Research & Development LLC, Beerse, Belgium
| |
Collapse
|
37
|
Sciarra A, Pintea B, Nahm JH, Donadon M, Morenghi E, Maggioni M, Blanc JF, Torzilli G, Yeh M, Bioulac-Sage P, Park YN, Roncalli M, Di Tommaso L. CYP1A2 is a predictor of HCC recurrence in HCV-related chronic liver disease: A retrospective multicentric validation study. Dig Liver Dis 2017; 49:434-439. [PMID: 28040498 DOI: 10.1016/j.dld.2016.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 11/30/2016] [Accepted: 12/05/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Although hepatic resection is a potentially curative treatment for hepatocellular carcinoma (HCC), post-operative prognosis remains unsatisfactory due to the high incidence of recurrence. Several clinicopathological markers have been associated with HCC recurrence, but none has been validated. Extratumoral expression of cytochrome P4501A2 (CYP1A2) was recently proposed as predictor of HCC recurrence. AIMS To validate extratumoral CYP1A2 as predictor of HCC recurrence and to determine its applicability to pretreatment liver biopsy. METHODS Surgically resected HCC (n.180) with clinicopathological data and follow up were retrospectively studied (HCV n.54; HBV n.91; NAFLD/NASH n.35). CYP1A2 expression was evaluated using an immunohistochemical assay and semiquantitative analysis. RESULTS Etiology-stratified analysis showed that low CYP1A2 expression was independently associated with recurrence-free survival in HCV patients (HR 2.814, 95% CI 1.300-6.093, p=0.009); this association was lost in the whole cohort. Pretreatment liver biopsy and paired surgical specimens showed concordant CYP1A2 expression in the vast majority of cases (87%), with NPV of 100%, PPV of 81.25%, and a Cohen kappa of 0.72 (substantial agreement). CONCLUSION We validated the extratumoral expression of CYP1A2 as a biomarker of HCC recurrence in HCV patients. CYP1A2 analysis in pretreatment liver biopsy can be of help to stratify HCC patients for personalized treatment.
Collapse
Affiliation(s)
| | - Bogdan Pintea
- University of Medicine and Pharmacy Iuliu Hatieganu, Cluj-Napoca, Romania
| | - Ji Hae Nahm
- Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Matteo Donadon
- Humanitas Clinical and Research Center, Hepatobiliary and General Surgery, Rozzano, Milano, Italy
| | - Emanuela Morenghi
- Humanitas Clinical and Research Center, Biostatistics Unit, Rozzano, Milano, Italy
| | - Marco Maggioni
- Pathology, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milan, Italy
| | - Jean Frederic Blanc
- Hepato-Gastroenterology and Digestive Oncology, Saint André Hospital CHU Bordeaux and Inserm UMR 1053, Bordeaux University, Bordeaux, France
| | - Guido Torzilli
- Humanitas Clinical and Research Center, Hepatobiliary and General Surgery, Rozzano, Milano, Italy; Humanitas University, Department of Biomedical Sciences, Rozzano, Milano, Italy
| | - Matthew Yeh
- Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Paulette Bioulac-Sage
- Pathology, CHU de Bordeaux, Pellegrin Hospital, Bordeaux, France and Inserm UMR 1053, Bordeaux University, Bordeaux, France
| | - Young Nyun Park
- Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Massimo Roncalli
- Humanitas Clinical and Research Center, Pathology Unit, Rozzano, Milano, Italy; Humanitas University, Department of Biomedical Sciences, Rozzano, Milano, Italy.
| | - Luca Di Tommaso
- Humanitas Clinical and Research Center, Pathology Unit, Rozzano, Milano, Italy; Humanitas University, Department of Biomedical Sciences, Rozzano, Milano, Italy.
| |
Collapse
|
38
|
Procalcitonin Impairs Liver Cell Viability and Function In Vitro: A Potential New Mechanism of Liver Dysfunction and Failure during Sepsis? BIOMED RESEARCH INTERNATIONAL 2017; 2017:6130725. [PMID: 28255555 PMCID: PMC5309405 DOI: 10.1155/2017/6130725] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Revised: 01/05/2017] [Accepted: 01/10/2017] [Indexed: 01/09/2023]
Abstract
Purpose. Liver dysfunction and failure are severe complications of sepsis and result in poor outcome and increased mortality. The underlying pathologic mechanisms of hepatocyte dysfunction and necrosis during sepsis are only incompletely understood. Here, we investigated whether procalcitonin, a biomarker of sepsis, modulates liver cell function and viability. Materials and Methods. Employing a previously characterized and patented biosensor system evaluating hepatocyte toxicity in vitro, human hepatocellular carcinoma cells (HepG2/C3A) were exposed to 0.01-50 ng/mL procalcitonin for 2 × 72 h and evaluated for proliferation, necrosis, metabolic activity, cellular integrity, microalbumin synthesis, and detoxification capacity. Acetaminophen served as positive control. For further standardization, procalcitonin effects were confirmed in a cellular toxicology assay panel employing L929 fibroblasts. Data were analyzed using ANOVA/Tukey's test. Results. Already at concentrations as low as 0.25 ng/mL, procalcitonin induced HepG2/C3A necrosis (P < 0.05) and reduced metabolic activity, cellular integrity, synthesis, and detoxification capacity (all P < 0.001). Comparable effects were obtained employing L929 fibroblasts. Conclusion. We provide evidence for procalcitonin to directly impair function and viability of human hepatocytes and exert general cytotoxicity in vitro. Therapeutical targeting of procalcitonin could thus display a novel approach to reduce incidence of liver dysfunction and failure during sepsis and lower morbidity and mortality of septic patients.
Collapse
|
39
|
Abstract
Despite the central role of the liver in drug metabolism, surprisingly there is lack of certainty in anticipating the extent of modification of the clearance of a given drug in a given patient. The intent of this review is to provide a conceptual framework in considering the impact of liver disease on drug disposition and reciprocally the impact of drug disposition on liver disease. It is proposed that improved understanding of the situation is gained by considering the issue as a special example of a drug-gene-environment interaction. This requires an integration of knowledge of the drug's properties, knowledge of the gene products involved in its metabolism, and knowledge of the pathophysiology of its disposition. This will enhance the level of predictability of drug disposition and toxicity for a drug of interest in an individual patient. It is our contention that advances in pharmacology, pharmacogenomics, and hepatology, together with concerted interests in the academic, regulatory, and pharmaceutical industry communities provide an ideal immediate environment to move from a qualitative reactive approach to quantitative proactive approach in individualizing patient therapy in liver disease.
Collapse
Affiliation(s)
- Nathalie K Zgheib
- a Department of Pharmacology and Toxicology , American University of Beirut Faculty of Medicine , Beirut , Lebanon
| | - Robert A Branch
- b Department of Medicine, School of Medicine , University of Pittsburgh , Pittsburgh , PA , USA
| |
Collapse
|
40
|
Ivanov AV, Valuev-Elliston VT, Tyurina DA, Ivanova ON, Kochetkov SN, Bartosch B, Isaguliants MG. Oxidative stress, a trigger of hepatitis C and B virus-induced liver carcinogenesis. Oncotarget 2017; 8:3895-3932. [PMID: 27965466 PMCID: PMC5354803 DOI: 10.18632/oncotarget.13904] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 12/05/2016] [Indexed: 12/11/2022] Open
Abstract
Virally induced liver cancer usually evolves over long periods of time in the context of a strongly oxidative microenvironment, characterized by chronic liver inflammation and regeneration processes. They ultimately lead to oncogenic mutations in many cellular signaling cascades that drive cell growth and proliferation. Oxidative stress, induced by hepatitis viruses, therefore is one of the factors that drives the neoplastic transformation process in the liver. This review summarizes current knowledge on oxidative stress and oxidative stress responses induced by human hepatitis B and C viruses. It focuses on the molecular mechanisms by which these viruses activate cellular enzymes/systems that generate or scavenge reactive oxygen species (ROS) and control cellular redox homeostasis. The impact of an altered cellular redox homeostasis on the initiation and establishment of chronic viral infection, as well as on the course and outcome of liver fibrosis and hepatocarcinogenesis will be discussed The review neither discusses reactive nitrogen species, although their metabolism is interferes with that of ROS, nor antioxidants as potential therapeutic remedies against viral infections, both subjects meriting an independent review.
Collapse
Affiliation(s)
- Alexander V. Ivanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | | | - Daria A. Tyurina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Olga N. Ivanova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Sergey N. Kochetkov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Birke Bartosch
- Inserm U1052, Cancer Research Center Lyon, University of Lyon, Lyon, France
- DevWeCan Laboratories of Excellence Network, France
| | - Maria G. Isaguliants
- Riga Stradins University, Riga, Latvia
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
41
|
Medvedev R, Hildt E, Ploen D. Look who's talking-the crosstalk between oxidative stress and autophagy supports exosomal-dependent release of HCV particles. Cell Biol Toxicol 2016; 33:211-231. [PMID: 27987184 DOI: 10.1007/s10565-016-9376-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 12/04/2016] [Indexed: 12/12/2022]
Abstract
Autophagy is a highly conserved and regulated intracellular lysosomal degradation pathway that is essential for cell survival. Dysregulation has been linked to the development of various human diseases, including neurodegeneration and tumorigenesis, infection, and aging. Besides, many viruses hijack the autophagosomal pathway to support their life cycle. The hepatitis C virus (HCV), a major cause of chronic liver diseases worldwide, has been described to induce autophagy. The autophagosomal pathway can be further activated in response to elevated levels of reactive oxygen species (ROS). HCV impairs the Nrf2/ARE-dependent induction of ROS-detoxifying enzymes by a so far unprecedented mechanism. In line with this, this review aims to discuss the relevance of HCV-dependent elevated ROS levels for the induction of autophagy as a result of the impaired Nrf2 signaling and the described crosstalk between p62 and the Nrf2/Keap1 signaling pathway. Moreover, autophagy is functionally connected to the endocytic pathway as components of the endosomal trafficking are involved in the maturation of autophagosomes. The release of HCV particles is still not fully understood. Recent studies suggest an involvement of exosomes that originate from the endosomal pathway in viral release. In line with this, it is tempting to speculate whether HCV-dependent elevated ROS levels induce autophagy to support exosome-mediated release of viral particles. Based on recent findings, in this review, we will further highlight the impact of HCV-induced autophagy and its interplay with the endosomal pathway as a novel mechanism for the release of HCV particles.
Collapse
Affiliation(s)
- Regina Medvedev
- Department of Virology, Paul-Ehrlich-Institut, Paul-Ehrlich-Straße 51-59, 63225, Langen, Germany
| | - Eberhard Hildt
- Department of Virology, Paul-Ehrlich-Institut, Paul-Ehrlich-Straße 51-59, 63225, Langen, Germany.,Deutsches Zentrum für Infektionsforschung (DZIF), Gießen, Marburg, Langen, Germany
| | - Daniela Ploen
- Department of Virology, Paul-Ehrlich-Institut, Paul-Ehrlich-Straße 51-59, 63225, Langen, Germany.
| |
Collapse
|
42
|
Ning J, Yang Z, Xie S, Sun Y, Yuan C, Chen H. Hepatic function imaging using dynamic Gd-EOB-DTPA enhanced MRI and pharmacokinetic modeling. Magn Reson Med 2016; 78:1488-1495. [PMID: 27785826 DOI: 10.1002/mrm.26520] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Revised: 09/17/2016] [Accepted: 09/28/2016] [Indexed: 12/18/2022]
Abstract
PURPOSE To determine whether pharmacokinetic modeling parameters with different output assumptions of dynamic contrast-enhanced MRI (DCE-MRI) using Gd-EOB-DTPA correlate with serum-based liver function tests, and compare the goodness of fit of the different output assumptions. METHODS A 6-min DCE-MRI protocol was performed in 38 patients. Four dual-input two-compartment models with different output assumptions and a published one-compartment model were used to calculate hepatic function parameters. The Akaike information criterion fitting error was used to evaluate the goodness of fit. Imaging-based hepatic function parameters were compared with blood chemistry using correlation with multiple comparison correction. RESULTS The dual-input two-compartment model assuming venous flow equals arterial flow plus portal venous flow and no bile duct output better described the liver tissue enhancement with low fitting error and high correlation with blood chemistry. The relative uptake rate Kir derived from this model was found to be significantly correlated with direct bilirubin (r = -0.52, P = 0.015), prealbumin concentration (r = 0.58, P = 0.015), and prothrombin time (r = -0.51, P = 0.026). CONCLUSION It is feasible to evaluate hepatic function by proper output assumptions. The relative uptake rate has the potential to serve as a biomarker of function. Magn Reson Med 78:1488-1495, 2017. © 2016 International Society for Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
- Jia Ning
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Zhiying Yang
- Department of Hepatobiliary Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Sheng Xie
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Yongliang Sun
- Department of Hepatobiliary Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Chun Yuan
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China.,Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Huijun Chen
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
43
|
Yang L, Ding Y, Rao S, Chen C, Wu L, Sheng R, Fu C, Zeng M. Staging liver fibrosis in chronic hepatitis B with T 1 relaxation time index on gadoxetic acid-enhanced MRI: Comparison with aspartate aminotransferase-to-platelet ratio index and FIB-4. J Magn Reson Imaging 2016; 45:1186-1194. [PMID: 27563840 DOI: 10.1002/jmri.25440] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 08/08/2016] [Indexed: 12/17/2022] Open
Abstract
PURPOSE To assess the accuracy of the T1 relaxation time index on gadoxetic acid-enhanced magnetic resonance imaging (MRI) for staging liver fibrosis in chronic hepatitis B (CHB), in comparison and combination with the aspartate aminotransferase-to-platelet ratio index (APRI) and fibrosis-4 (FIB-4). MATERIALS AND METHODS A retrospective study of gadoxetic acid-enhanced T1 mapping and serum biochemical tests was performed on 126 CHB patients who underwent gadoxetic acid-enhanced 1.5T MRI, and the histological score used as the gold standard. The reduction rate of T1 relaxation time before and 20 minutes after gadoxetic acid injection (ΔT1 , ΔR1%), the contrast uptake rate (KHep ), APRI, and FIB-4 were calculated. The diagnostic efficacy of ΔT1 , ΔR1%, KHep , APRI, and FIB-4 for predicting stage 2 or greater (≥S2), stage 3 or greater (≥S3), and stage 4 (S4) was compared. RESULTS ΔT1 (r = -0.513, P < 0.001), ΔR1% (r = -0.626, P < 0.001), KHep (r = -0.527, P < 0.001), APRI (r = 0.519, P < 0.001), and FIB-4 (r = 0.476, P < 0.001) correlated significantly with fibrosis stages. Areas under the curves (AUCs) of ΔR1% for detecting ≥S2, ≥S3, and S4 were 0.849, 0.827, and 0.809, which were greater than that of APRI (0.763, 0.745, 0.787) and FIB-4 (0.727, 0.738, 0.772), but significant difference was found only in discriminating ≥S2 between ΔR1% and FIB-4 (P = 0.027). The combination of all five indices performed best, with AUC, sensitivity, and specificity of 0.860, 87.21%, and 72.50% for diagnosing ≥S2, 0.878, 82.81%, and 85.48% for ≥S3, and 0.867, 80.00%, and 83.95% for S4. CONCLUSION The gadoxetic acid-enhanced T1 relaxation time index appears to be superior to APRI and FIB-4 for predicting hepatic fibrosis. The combined use of gadoxetic acid-enhanced T1 mapping, APRI, and FIB-4 may be more reliable for staging liver fibrosis in CHB. LEVEL OF EVIDENCE 4 J. Magn. Reson. Imaging 2017;45:1186-1194.
Collapse
Affiliation(s)
- Li Yang
- Department of Radiology, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Ying Ding
- Department of Radiology, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, P.R. China.,Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Shengxiang Rao
- Department of Radiology, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, P.R. China.,Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Caizhong Chen
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Lifang Wu
- Department of Radiology, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Ruofan Sheng
- Department of Radiology, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, P.R. China.,Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Caixia Fu
- Siemens Healthcare, Shanghai, P.R. China
| | - Mengsu Zeng
- Department of Radiology, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, P.R. China.,Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| |
Collapse
|
44
|
Bioartificial Therapy of Sepsis: Changes of Norepinephrine-Dosage in Patients and Influence on Dynamic and Cell Based Liver Tests during Extracorporeal Treatments. BIOMED RESEARCH INTERNATIONAL 2016; 2016:7056492. [PMID: 27433475 PMCID: PMC4940519 DOI: 10.1155/2016/7056492] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 06/02/2016] [Indexed: 01/28/2023]
Abstract
Purpose. Granulocyte transfusions have been used to treat immune cell dysfunction in sepsis. A granulocyte bioreactor for the extracorporeal treatment of sepsis was tested in a prospective clinical study focusing on the dosage of norepinephrine in patients and influence on dynamic and cell based liver tests during extracorporeal therapies. Methods and Patients. Ten patients with severe sepsis were treated twice within 72 h with the system containing granulocytes from healthy donors. Survival, physiologic parameters, extended hemodynamic measurement, and the indocyanine green plasma disappearance rate (PDR) were monitored. Plasma of patients before and after extracorporeal treatments were tested with a cell based biosensor for analysis of hepatotoxicity. Results. The observed mortality rate was 50% during stay in hospital. During the treatments, the norepinephrine-dosage could be significantly reduced while mean arterial pressure was stable. In the cell based analysis of hepatotoxicity, the viability and function of sensor-cells increased significantly during extracorporeal treatment in all patients and the PDR-values increased significantly between day 1 and day 7 only in survivors. Conclusion. The extracorporeal treatment with donor granulocytes showed promising effects on dosage of norepinephrine in patients, liver cell function, and viability in a cell based biosensor. Further studies with this approach are encouraged.
Collapse
|
45
|
Kang J, Wang J, Cheng J, Cao Z, Chen R, Li H, Liu S, Chen X, Sui J, Lu F. Down-regulation of NTCP expression by cyclin D1 in hepatitis B virus-related hepatocellular carcinoma has clinical significance. Oncotarget 2016; 8:56041-56050. [PMID: 28915572 PMCID: PMC5593543 DOI: 10.18632/oncotarget.10241] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 05/29/2016] [Indexed: 12/22/2022] Open
Abstract
The sodium-dependent taurocholate cotransporter polypeptide (NTCP) has been identified as a liver specific functional receptor for the hepatitis B virus (HBV). Previous studies indicated that the expression of NTCP may be associated with the proliferation status of hepatocytes. However, the involvement of NTCP in hepatocellular carcinoma (HCC) cells proliferation remains unclear. In this study, we confirmed that NTCP was down-regulated in HCC tumor tissues compared with that in the adjacent non-tumor tissues (P < 0.0001). Clinically, lower expression of NTCP was correlated with poor post-surgery survival rate (P = 0.0009) and larger tumor tissue mass (P = 0.003) of HCC patients. This was supported by the finding that ectopic expression of NTCP in both HepG2 and Huh-7 cells could significantly suppress hepatocytes growth by arresting cells in G0/G1 phase. We also discovered that cyclin D1 could transcriptionally suppress NTCP expression by inhibiting the activity of NTCP promoter, while arresting HCC cells in G0/G1 phase by serum starvation could upregulate NTCP mRNA levels. This is the first study to report that the transcriptional inhibition of NTCP expression during cell cycle progression was mediated by cyclin D1. The down-regulated NTCP expression was associated with poor prognosis and lower HBV cccDNA level in HCC patients. Therefore, NTCP expression levels might serve as a novel prognostic predictive marker for post-surgery survival rate of HCC patients.
Collapse
Affiliation(s)
- Jingting Kang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Science, Peking University Health Science Center, Beijing 100191, P. R. China
| | - Jie Wang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Science, Peking University Health Science Center, Beijing 100191, P. R. China
| | - Jin Cheng
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Science, Peking University Health Science Center, Beijing 100191, P. R. China
| | - Zhiliang Cao
- National Institute of Biological Sciences, Zhongguancun Life Science Park, Changping, Beijing, 102206, China
| | - Ran Chen
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Science, Peking University Health Science Center, Beijing 100191, P. R. China
| | - Huiyu Li
- National Institute of Biological Sciences, Zhongguancun Life Science Park, Changping, Beijing, 102206, China
| | - Shuang Liu
- Beijing Artificial Liver Treatment and Training Center, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, P.R. China
| | - Xiangmei Chen
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Science, Peking University Health Science Center, Beijing 100191, P. R. China
| | - Jianhua Sui
- National Institute of Biological Sciences, Zhongguancun Life Science Park, Changping, Beijing, 102206, China
| | - Fengmin Lu
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Science, Peking University Health Science Center, Beijing 100191, P. R. China
| |
Collapse
|
46
|
Biosensor for Hepatocellular Injury Corresponds to Experimental Scoring of Hepatosplenic Schistosomiasis in Mice. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1567254. [PMID: 27376078 PMCID: PMC4916270 DOI: 10.1155/2016/1567254] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 05/17/2016] [Indexed: 12/18/2022]
Abstract
Severe hepatosplenic injury of mansonian schistosomiasis is caused by Th2 mediated granulomatous response against parasite eggs entrapped within the periportal tissue. Subsequent fibrotic scarring and deformation/sclerosing of intrahepatic portal veins lead to portal hypertension, ascites, and oesophageal varices. The murine model of Schistosoma mansoni (S. mansoni) infection is suitable to establish the severe hepatosplenic injury of disease within a reasonable time scale for the development of novel antifibrotic or anti-infective strategies against S. mansoni infection. The drawback of the murine model is that the material prepared for complex analysis of egg burden, granuloma size, hepatic inflammation, and fibrosis is limited due to small amounts of liver tissue and blood samples. The objective of our study was the implementation of a macroscopic scoring system for mice livers to determine infection-related organ alterations of S. mansoni infection. In addition, an in vitro biosensor system based on the detection of hepatocellular injury in HepG2/C3A cells following incubation with serum of moderately (50 S. mansoni cercariae) and heavily (100 S. mansoni cercariae) infected mice affirmed the value of our scoring system. Therefore, our score represents a valuable tool in experimental schistosomiasis to assess severity of hepatosplenic schistosomiasis and reduce animal numbers by saving precious tissue samples.
Collapse
|
47
|
Baba H, Tajiri K, Nagata K, Kawai K, Minemura M, Sugiyama T. Hyperbilirubinemia without Transaminitis during Combined Therapy with Daclatasvir and Asunaprevir. Case Rep Gastroenterol 2016; 10:352-359. [PMID: 27504082 PMCID: PMC4965544 DOI: 10.1159/000447486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 06/07/2016] [Indexed: 02/05/2023] Open
Abstract
Daclatasvir (DCV) and asunaprevir (ASV) are direct-acting antivirals (DAAs) used in the treatment of chronic hepatitis C virus (HCV) infection. Combined therapy with DCV and ASV shows high efficacy and safety even in patients with cirrhosis. We encountered a patient exhibiting severe hyperbilirubinemia during combined therapy, which is an unreported side effect of DCV and ASV. A 78-year-old woman with cirrhosis developed hyperbilirubinemia >10 mg/dl without transaminitis 3 weeks after starting combined therapy. We suspected DAAs-induced liver disorder and discontinued treatment, which resulted in the improvement of hyperbilirubinemia. Caution is required in the use of DAAs for patients with advanced cirrhosis.
Collapse
Affiliation(s)
- Hayato Baba
- The Third Department of Internal Medicine, Toyama University Hospital, Toyama, Japan
- Department of Education and Clinical Training, Toyama University Hospital, Toyama, Japan
| | - Kazuto Tajiri
- The Third Department of Internal Medicine, Toyama University Hospital, Toyama, Japan
| | - Kohei Nagata
- The Third Department of Internal Medicine, Toyama University Hospital, Toyama, Japan
| | - Kengo Kawai
- The Third Department of Internal Medicine, Toyama University Hospital, Toyama, Japan
| | - Masami Minemura
- The Third Department of Internal Medicine, Toyama University Hospital, Toyama, Japan
| | - Toshiro Sugiyama
- The Third Department of Internal Medicine, Toyama University Hospital, Toyama, Japan
| |
Collapse
|
48
|
Smirnova OA, Ivanova ON, Bartosch B, Valuev-Elliston VT, Mukhtarov F, Kochetkov SN, Ivanov AV. Hepatitis C Virus NS5A Protein Triggers Oxidative Stress by Inducing NADPH Oxidases 1 and 4 and Cytochrome P450 2E1. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:8341937. [PMID: 27200149 PMCID: PMC4855014 DOI: 10.1155/2016/8341937] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 02/03/2016] [Accepted: 03/01/2016] [Indexed: 02/06/2023]
Abstract
Replication of hepatitis C virus (HCV) is associated with the induction of oxidative stress, which is thought to play a major role in various liver pathologies associated with chronic hepatitis C. NS5A protein of the virus is one of the two key viral proteins that are known to trigger production of reactive oxygen species (ROS). To date it has been considered that NS5A induces oxidative stress by altering calcium homeostasis. Herein we show that NS5A-induced oxidative stress was only moderately inhibited by the intracellular calcium chelator BAPTA-AM and not at all inhibited by the drug that blocks the Ca(2+) flux from ER to mitochondria. Furthermore, ROS production was not accompanied by induction of ER oxidoreductins (Ero1), H2O2-producing enzymes that are implicated in the regulation of calcium fluxes. Instead, we found that NS5A contributes to ROS production by activating expression of NADPH oxidases 1 and 4 as well as cytochrome P450 2E1. These effects were mediated by domain I of NS5A protein. NOX1 and NOX4 induction was mediated by enhanced production of transforming growth factor β1 (TGFβ1). Thus, our data show that NS5A protein induces oxidative stress by several multistep mechanisms.
Collapse
Affiliation(s)
- Olga A. Smirnova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Street 32, Moscow 119991, Russia
| | - Olga N. Ivanova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Street 32, Moscow 119991, Russia
| | - Birke Bartosch
- CRCL, INSERM U1052, CNRS 5286, Université de Lyon, 151 Cours A. Thomas, 69424 Lyon Cedex, France
| | - Vladimir T. Valuev-Elliston
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Street 32, Moscow 119991, Russia
| | - Furkat Mukhtarov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Street 32, Moscow 119991, Russia
| | - Sergey N. Kochetkov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Street 32, Moscow 119991, Russia
| | - Alexander V. Ivanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Street 32, Moscow 119991, Russia
| |
Collapse
|
49
|
HCV and Oxidative Stress: Implications for HCV Life Cycle and HCV-Associated Pathogenesis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:9012580. [PMID: 26955431 PMCID: PMC4756209 DOI: 10.1155/2016/9012580] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 01/14/2016] [Indexed: 12/15/2022]
Abstract
HCV (hepatitis C virus) is a member of the Flaviviridae family that contains a single-stranded positive-sense RNA genome of approximately 9600 bases. HCV is a major causative agent for chronic liver diseases such as steatosis, fibrosis, cirrhosis, and hepatocellular carcinoma which are caused by multifactorial processes. Elevated levels of reactive oxygen species (ROS) are considered as a major factor contributing to HCV-associated pathogenesis. This review summarizes the mechanisms involved in formation of ROS in HCV replicating cells and describes the interference of HCV with ROS detoxifying systems. The relevance of ROS for HCV-associated pathogenesis is reviewed with a focus on the interference of elevated ROS levels with processes controlling liver regeneration. The overview about the impact of ROS for the viral life cycle is focused on the relevance of autophagy for the HCV life cycle and the crosstalk between HCV, elevated ROS levels, and the induction of autophagy.
Collapse
|
50
|
Dietrich CG, Götze O, Geier A. Molecular changes in hepatic metabolism and transport in cirrhosis and their functional importance. World J Gastroenterol 2016; 22:72-88. [PMID: 26755861 PMCID: PMC4698509 DOI: 10.3748/wjg.v22.i1.72] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 09/24/2015] [Accepted: 11/13/2015] [Indexed: 02/06/2023] Open
Abstract
Liver cirrhosis is the common endpoint of many hepatic diseases and represents a relevant risk for liver failure and hepatocellular carcinoma. The progress of liver fibrosis and cirrhosis is accompanied by deteriorating liver function. This review summarizes the regulatory and functional changes in phase I and phase II metabolic enzymes as well as transport proteins and provides an overview regarding lipid and glucose metabolism in cirrhotic patients. Interestingly, phase I enzymes are generally downregulated transcriptionally, while phase II enzymes are mostly preserved transcriptionally but are reduced in their function. Transport proteins are regulated in a specific way that resembles the molecular changes observed in obstructive cholestasis. Lipid and glucose metabolism are characterized by insulin resistance and catabolism, leading to the disturbance of energy expenditure and wasting. Possible non-invasive tests, especially breath tests, for components of liver metabolism are discussed. The heterogeneity and complexity of changes in hepatic metabolism complicate the assessment of liver function in individual patients. Additionally, studies in humans are rare, and species differences preclude the transferability of data from rodents to humans. In clinical practice, some established global scores or criteria form the basis for the functional evaluation of patients with liver cirrhosis, but difficult treatment decisions such as selection for transplantation or resection require further research regarding the application of existing non-invasive tests and the development of more specific tests.
Collapse
|