201
|
Nasr P, Jönsson C, Ekstedt M, Kechagias S. Non-metabolic causes of steatotic liver disease. METABOLISM AND TARGET ORGAN DAMAGE 2023; 3. [DOI: 10.20517/mtod.2023.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Hepatic steatosis is caused by exaggerated hepatic lipid accumulation and is a common histological and radiological finding. Non-alcoholic fatty liver disease (NAFLD), or metabolic dysfunction associated steatotic liver disease (MASLD), is highly associated with metabolic syndrome and represents the most common cause of hepatic steatosis. However, since several comorbidities, lifestyle factors, and drugs can cause hepatic steatosis, MASLD is, to some extent, a diagnosis of exclusion. Nevertheless, initiatives have been taken to encompass positive (instead of negative) criteria for diagnosis - such as the presence of cardiometabolic risk factors together with hepatic steatosis. Nonetheless, before confirming a patient with MASLD, it is essential to map and evaluate other causes of fatty liver disease or steatotic liver disease. Several causes of hepatic steatosis have been identified in studies; however, the study cohorts are scarce and often anecdotal. Additionally, many studies have shown correlation without proving causation, and many are retrospective without reporting relevant patient characteristics and comorbidities - making it difficult to draw conclusions regarding the underlying etiology or present comorbidity of hepatic steatosis. In this narrative review, we aimed to identify and summarize present studies evaluating the impact of the most common and often suggested causes of hepatic steatosis.
Collapse
|
202
|
Bernasconi E, Biagi M, Di Agostino S, Cursaro C, Felicani C, Ronconi E, Franchi E, Costanzo AC, Gabrielli F, Cavicchioli A, Ienopoli G, Marenghi P, Bartoli A, Serra B, Scalabrini D, Sighinolfi P, Andreone P. Investigating Acute Hepatitis after SARS-CoV-2 Vaccination or Infection: A Genetic Case Series. Biomedicines 2023; 11:2848. [PMID: 37893221 PMCID: PMC10604753 DOI: 10.3390/biomedicines11102848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/14/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
(1) Background: Despite the advantages of COVID-19 vaccination, rare cases of acute hepatitis developing after the administration of the COVID-19 vaccine or the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection have been reported. The aim of the study is to describe a case series of patients who experienced the onset of acute hepatitis, with or without autoimmune features, following SARS-CoV-2 vaccination or infection and to hypothesize a genetic susceptibility in the pathogenesis. (2) Methods: A group of patients with acute onset hepatitis following SARS-CoV-2 vaccination or infection were evaluated in our hepatology outpatient clinic, where they underwent biochemical and autoimmune tests. Hepatitis A (HAV), B (HBV), and C virus (HCV), cytomegalovirus (CMV), Epstein-Barr virus (EBV), and human immunodeficiency virus (HIV) infections were excluded. Patients with a diagnosis of autoimmune hepatitis (AIH) or drug-induced liver injury (DILI) underwent HLA typing and histological testing. (3) Results: Five patients experienced new-onset AIH after COVID-19 vaccination, one of which developed mild symptoms after vaccination that strongly worsened during subsequent SARS-CoV-2 infection. One patient had AIH relapse after COVID-19 vaccination while on maintenance immunosuppressive treatment. All of them had HLA DRB1 alleles known to confer susceptibility to AIH (HLA DRB1*03,*07,*13,*14), and in three of them, HLA DRB1*11 was also detected. Two patients developed acute hepatitis without autoimmune hallmarks which resolved spontaneously, both positive for HLA DRB1*11. (4) Conclusions: An association between AIH and COVID-19 vaccine or infection can be hypothesized in individuals with a genetic predisposition. In patients without autoimmune features and spontaneous improvement of hypertransaminasemia, the diagnosis of drug-induced liver injury (DILI) is probable. Further studies are needed to determine the presence of an actual association and identify a possible role of HLA DRB1*11 in the pathogenesis of acute liver injury after SARS-CoV2 vaccination or infection.
Collapse
Affiliation(s)
- Elisa Bernasconi
- Department of Internal Medicine, Civil Hospital of Baggiovara, University of Modena and Reggio Emilia, Baggiovara, 41126 Modena, Italy; (E.B.); (M.B.); (S.D.A.); (C.C.); (C.F.); (E.R.); (E.F.); (A.C.C.); (F.G.); (A.C.); (G.I.); (P.M.); (A.B.); (B.S.); (D.S.); (P.S.)
| | - Matteo Biagi
- Department of Internal Medicine, Civil Hospital of Baggiovara, University of Modena and Reggio Emilia, Baggiovara, 41126 Modena, Italy; (E.B.); (M.B.); (S.D.A.); (C.C.); (C.F.); (E.R.); (E.F.); (A.C.C.); (F.G.); (A.C.); (G.I.); (P.M.); (A.B.); (B.S.); (D.S.); (P.S.)
| | - Stefania Di Agostino
- Department of Internal Medicine, Civil Hospital of Baggiovara, University of Modena and Reggio Emilia, Baggiovara, 41126 Modena, Italy; (E.B.); (M.B.); (S.D.A.); (C.C.); (C.F.); (E.R.); (E.F.); (A.C.C.); (F.G.); (A.C.); (G.I.); (P.M.); (A.B.); (B.S.); (D.S.); (P.S.)
| | - Carmela Cursaro
- Department of Internal Medicine, Civil Hospital of Baggiovara, University of Modena and Reggio Emilia, Baggiovara, 41126 Modena, Italy; (E.B.); (M.B.); (S.D.A.); (C.C.); (C.F.); (E.R.); (E.F.); (A.C.C.); (F.G.); (A.C.); (G.I.); (P.M.); (A.B.); (B.S.); (D.S.); (P.S.)
| | - Cristina Felicani
- Department of Internal Medicine, Civil Hospital of Baggiovara, University of Modena and Reggio Emilia, Baggiovara, 41126 Modena, Italy; (E.B.); (M.B.); (S.D.A.); (C.C.); (C.F.); (E.R.); (E.F.); (A.C.C.); (F.G.); (A.C.); (G.I.); (P.M.); (A.B.); (B.S.); (D.S.); (P.S.)
| | - Enrico Ronconi
- Department of Internal Medicine, Civil Hospital of Baggiovara, University of Modena and Reggio Emilia, Baggiovara, 41126 Modena, Italy; (E.B.); (M.B.); (S.D.A.); (C.C.); (C.F.); (E.R.); (E.F.); (A.C.C.); (F.G.); (A.C.); (G.I.); (P.M.); (A.B.); (B.S.); (D.S.); (P.S.)
| | - Elena Franchi
- Department of Internal Medicine, Civil Hospital of Baggiovara, University of Modena and Reggio Emilia, Baggiovara, 41126 Modena, Italy; (E.B.); (M.B.); (S.D.A.); (C.C.); (C.F.); (E.R.); (E.F.); (A.C.C.); (F.G.); (A.C.); (G.I.); (P.M.); (A.B.); (B.S.); (D.S.); (P.S.)
| | - Arianna Carmen Costanzo
- Department of Internal Medicine, Civil Hospital of Baggiovara, University of Modena and Reggio Emilia, Baggiovara, 41126 Modena, Italy; (E.B.); (M.B.); (S.D.A.); (C.C.); (C.F.); (E.R.); (E.F.); (A.C.C.); (F.G.); (A.C.); (G.I.); (P.M.); (A.B.); (B.S.); (D.S.); (P.S.)
| | - Filippo Gabrielli
- Department of Internal Medicine, Civil Hospital of Baggiovara, University of Modena and Reggio Emilia, Baggiovara, 41126 Modena, Italy; (E.B.); (M.B.); (S.D.A.); (C.C.); (C.F.); (E.R.); (E.F.); (A.C.C.); (F.G.); (A.C.); (G.I.); (P.M.); (A.B.); (B.S.); (D.S.); (P.S.)
| | - Alessia Cavicchioli
- Department of Internal Medicine, Civil Hospital of Baggiovara, University of Modena and Reggio Emilia, Baggiovara, 41126 Modena, Italy; (E.B.); (M.B.); (S.D.A.); (C.C.); (C.F.); (E.R.); (E.F.); (A.C.C.); (F.G.); (A.C.); (G.I.); (P.M.); (A.B.); (B.S.); (D.S.); (P.S.)
| | - Giuseppe Ienopoli
- Department of Internal Medicine, Civil Hospital of Baggiovara, University of Modena and Reggio Emilia, Baggiovara, 41126 Modena, Italy; (E.B.); (M.B.); (S.D.A.); (C.C.); (C.F.); (E.R.); (E.F.); (A.C.C.); (F.G.); (A.C.); (G.I.); (P.M.); (A.B.); (B.S.); (D.S.); (P.S.)
| | - Paolo Marenghi
- Department of Internal Medicine, Civil Hospital of Baggiovara, University of Modena and Reggio Emilia, Baggiovara, 41126 Modena, Italy; (E.B.); (M.B.); (S.D.A.); (C.C.); (C.F.); (E.R.); (E.F.); (A.C.C.); (F.G.); (A.C.); (G.I.); (P.M.); (A.B.); (B.S.); (D.S.); (P.S.)
| | - Alessandra Bartoli
- Department of Internal Medicine, Civil Hospital of Baggiovara, University of Modena and Reggio Emilia, Baggiovara, 41126 Modena, Italy; (E.B.); (M.B.); (S.D.A.); (C.C.); (C.F.); (E.R.); (E.F.); (A.C.C.); (F.G.); (A.C.); (G.I.); (P.M.); (A.B.); (B.S.); (D.S.); (P.S.)
| | - Beatrice Serra
- Department of Internal Medicine, Civil Hospital of Baggiovara, University of Modena and Reggio Emilia, Baggiovara, 41126 Modena, Italy; (E.B.); (M.B.); (S.D.A.); (C.C.); (C.F.); (E.R.); (E.F.); (A.C.C.); (F.G.); (A.C.); (G.I.); (P.M.); (A.B.); (B.S.); (D.S.); (P.S.)
| | - Davide Scalabrini
- Department of Internal Medicine, Civil Hospital of Baggiovara, University of Modena and Reggio Emilia, Baggiovara, 41126 Modena, Italy; (E.B.); (M.B.); (S.D.A.); (C.C.); (C.F.); (E.R.); (E.F.); (A.C.C.); (F.G.); (A.C.); (G.I.); (P.M.); (A.B.); (B.S.); (D.S.); (P.S.)
| | - Pamela Sighinolfi
- Department of Internal Medicine, Civil Hospital of Baggiovara, University of Modena and Reggio Emilia, Baggiovara, 41126 Modena, Italy; (E.B.); (M.B.); (S.D.A.); (C.C.); (C.F.); (E.R.); (E.F.); (A.C.C.); (F.G.); (A.C.); (G.I.); (P.M.); (A.B.); (B.S.); (D.S.); (P.S.)
| | - Pietro Andreone
- Department of Internal Medicine, Civil Hospital of Baggiovara, University of Modena and Reggio Emilia, Baggiovara, 41126 Modena, Italy; (E.B.); (M.B.); (S.D.A.); (C.C.); (C.F.); (E.R.); (E.F.); (A.C.C.); (F.G.); (A.C.); (G.I.); (P.M.); (A.B.); (B.S.); (D.S.); (P.S.)
- Department of Internal Medicine, General, Emergency and Post-Acute, Division of Metabolic Internal Medicine, Civil Hospital of Baggiovara, Azienda Ospedaliero-Universitaria di Modena, Baggiovara, 41126 Modena, Italy
| |
Collapse
|
203
|
Maev IV, Polunina TE. [Drug-induced liver injury: diagnosis of exclusion]. TERAPEVT ARKH 2023; 95:611-620. [PMID: 38158894 DOI: 10.26442/00403660.2023.08.202329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 10/08/2023] [Indexed: 01/03/2024]
Abstract
Drug-induced liver injury (DILI) is a relevant issue in clinical practice and is still a diagnosis of exclusion. Despite the low incidence in the general population, DILI is the cause of most cases of acute hepatic injury and has a mortality rate of up to 50%. Despite many reports in the medical literature about the DILI mechanisms, a clear causal relationship between them, drugs, and risk factors has not been established. Current clinical practice is based on a combination of a thorough study of a history of risk factors, the timing of drug and dietary supplements' administration, and the analysis of laboratory and instrumental tests. It aligns with the international criteria of the Rousell Uclaf Causality Assessment Method (RUCAM), which is considered one of the main diagnostic algorithms for DILI. The article addresses current DILI classification, risk factors, diagnostic algorithms, causalities, clinical evaluation, promising liver function biomarkers, and specific treatment.
Collapse
Affiliation(s)
- I V Maev
- Yevdokimov Moscow State University of Medicine and Dentistry
| | - T E Polunina
- Yevdokimov Moscow State University of Medicine and Dentistry
| |
Collapse
|
204
|
Zhang ML, Li WX, Wang XY, Zhang H, Wu YL, Yang LQ, Chen XF, Zhang SQ, Chen YL, Feng KR, Tang JF. A gene expression profile-based approach to screen the occurrence and predisposed host characteristics of drug-induced liver injury: a case study of Psoralea corylifolia Linn. Front Chem 2023; 11:1259569. [PMID: 37867998 PMCID: PMC10588485 DOI: 10.3389/fchem.2023.1259569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 09/12/2023] [Indexed: 10/24/2023] Open
Abstract
Drug-induced liver injury (DILI) is one of the most common causes of a drug being withdrawn, and identifying the culprit drugs and the host factors at risk of causing DILI has become a current challenge. Recent studies have found that immune status plays a considerable role in the development of DILI. In this study, DILI-related differentially expressed genes mediated by immunoinflammatory cytokines were obtained from the Gene Expression Omnibus (GEO) database to predict the occurrence of DILI (named the DILI predictive gene set, DILI_PGS), and the predictability of the DILI_PGS was verified using the Connectivity Map (CMap) and LiverTox platforms. The results obtained DILI_PGS from the GEO database could predict 81.25% of liver injury drugs. In addition, the Coexpedia platform was used to predict the DILI_PGS-related characteristics of common host diseases and found that the DILI_PGS mainly involved immune-related diseases and tumor-related diseases. Then, animal models of immune stress (IS) and immunosuppressive (IP) were selected to simulate the immune status of the above diseases. Meanwhile, psoralen, a main component derived from Psoralea corylifolia Linn. with definite hepatotoxicity, was selected as an experimental drug with highly similar molecular fingerprints to three idiosyncratic hepatotoxic drugs (nefazodone, trovafloxacin, and nimesulide) from the same DILI_PGS dataset. The animal experiment results found a single administration of psoralen could significantly induce liver injury in IS mice, while there was no obvious liver function change in IP mice by repeatedly administering the same dose of psoralen, and the potential mechanism of psoralen-induced liver injury in IS mice may be related to regulating the expression of the TNF-related pathway. In conclusion, this study constructed the DILI_PGS with high accuracy to predict the occurrence of DILI and preliminarily identified the characteristics of host factors inducing DILI.
Collapse
Affiliation(s)
- Ming-Liang Zhang
- The Department of Pharmacy, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, China
| | - Wei-Xia Li
- The Department of Pharmacy, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, China
| | - Xiao-Yan Wang
- The Department of Pharmacy, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, China
| | - Hui Zhang
- The Department of Pharmacy, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, China
| | - Ya-Li Wu
- The Department of Pharmacy, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, China
| | - Liu-Qing Yang
- The Department of Pharmacy, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, China
| | - Xiao-Fei Chen
- The Department of Pharmacy, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, China
| | - Shu-Qi Zhang
- The Department of Pharmacy, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, China
| | - Yu-Long Chen
- Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Ke-Ran Feng
- The Department of Pharmacy, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, China
| | - Jin-Fa Tang
- The Department of Pharmacy, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, China
- Henan University of Traditional Chinese Medicine, Zhengzhou, China
| |
Collapse
|
205
|
Zhou W, He H, Wei Q, Che L, Zhao X, Liu W, Yan Y, Hu L, Du Y, Yin Z, Shuai Y, Yang L, Feng R. Puerarin protects against acetaminophen-induced oxidative damage in liver through activation of the Keap1/Nrf2 signaling pathway. Food Sci Nutr 2023; 11:6604-6615. [PMID: 37823166 PMCID: PMC10563760 DOI: 10.1002/fsn3.3609] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 07/08/2023] [Accepted: 07/22/2023] [Indexed: 10/13/2023] Open
Abstract
Puerarin (Pue) is a kind of isoflavone compound extracted from Pueraria lobata, which has significant antioxidant activity. Excessive use of acetaminophen (APAP) can cause oxidative stress in the liver and eventually lead to acute liver injury. The purpose of this study was to investigate the protective effect and the mechanism of puerarin on APAP-induced liver oxidative damage. In in vitro experiments, puerarin significantly increased the cell activity of HepG2 cells, reduced the ROS accumulation, alleviated the oxidative damage and mitochondrial dysfunction. In in vivo studies, our results showed that puerarin enhanced antioxidant activity and alleviated histopathological damage. Further studies showed that puerarin decreased the expression of Keap1, promoted the nuclear migration of Nrf2, and up-regulated the expression of GCLC, GCLM, HO-1 and NQO1. This study demonstrated that puerarin can protect APAP-induced liver injury via alleviating oxidative stress and mitochondrial dysfunction by affecting the nuclear migration of Nrf2 via inhibiting Keap1.
Collapse
Affiliation(s)
- Wanhai Zhou
- Sichuan Oil Cinnamon Engineering Technology Research CenterYibin UniversityYibinChina
- Faculty of Agriculture, Forestry and Food EngineeringYiBin UniversityYibinChina
| | - Heng He
- Natural Medicine Research Center, College of Veterinary MedicineSichuan Agricultural UniversityChengduChina
| | - Qin Wei
- Sichuan Oil Cinnamon Engineering Technology Research CenterYibin UniversityYibinChina
| | - Litao Che
- Sichuan Oil Cinnamon Engineering Technology Research CenterYibin UniversityYibinChina
- Faculty of Agriculture, Forestry and Food EngineeringYiBin UniversityYibinChina
| | - Xin Zhao
- Sichuan Oil Cinnamon Engineering Technology Research CenterYibin UniversityYibinChina
- Faculty of Agriculture, Forestry and Food EngineeringYiBin UniversityYibinChina
| | - Wenwen Liu
- Sichuan Oil Cinnamon Engineering Technology Research CenterYibin UniversityYibinChina
- Faculty of Agriculture, Forestry and Food EngineeringYiBin UniversityYibinChina
| | - Yue Yan
- Sichuan Oil Cinnamon Engineering Technology Research CenterYibin UniversityYibinChina
- Faculty of Agriculture, Forestry and Food EngineeringYiBin UniversityYibinChina
| | - Lianqing Hu
- Sichuan Oil Cinnamon Engineering Technology Research CenterYibin UniversityYibinChina
- Faculty of Agriculture, Forestry and Food EngineeringYiBin UniversityYibinChina
| | - Yonghua Du
- Sichuan Oil Cinnamon Engineering Technology Research CenterYibin UniversityYibinChina
- Faculty of Agriculture, Forestry and Food EngineeringYiBin UniversityYibinChina
| | - Zhongqiong Yin
- Natural Medicine Research Center, College of Veterinary MedicineSichuan Agricultural UniversityChengduChina
| | - Yongkang Shuai
- Sichuan Oil Cinnamon Engineering Technology Research CenterYibin UniversityYibinChina
| | - Li Yang
- Sichuan Oil Cinnamon Engineering Technology Research CenterYibin UniversityYibinChina
| | - Ruizhang Feng
- Sichuan Oil Cinnamon Engineering Technology Research CenterYibin UniversityYibinChina
| |
Collapse
|
206
|
Oddie PD, Heskin J, Leung S, Naous N, Garvey L, Cohen CE. Drug-induced liver injury secondary to increased levonorgestrel exposure in a patient taking ritonavir. Int J STD AIDS 2023; 34:903-905. [PMID: 37449366 DOI: 10.1177/09564624231185889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
We report the first published case of a drug induced liver injury (DILI) presumed secondary to a drug-drug interaction between ritonavir and levonorgestrel progestogen-only emergency contraception (POEC). Our patient is a 25-year-old female living with human immunodeficiency virus (HIV), taking antiretroviral therapy (ART) containing tenofovir alafenamide/emtricitabine and darunavir/ritonavir. She was found to have elevated transaminases at a routine clinic appointment consistent with hepatocellular DILI. Further investigation found the most likely cause of this was a drug-drug interaction (DDI) between the ritonavir component of her ART and recent use of levonorgestrel POEC 3 days earlier. Evidence suggests that ritonavir increases levonorgestrel exposure, yet our patient received double the usual dose as per dispensing guidance at the time. We review the pharmacokinetics of ritonavir-levonorgestrel DDIs and highlight the need for consistent guidelines on this topic.
Collapse
Affiliation(s)
- Philip D Oddie
- GUM/HIV Directorate, Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| | - Joseph Heskin
- GUM/HIV Directorate, Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| | - Suki Leung
- GUM/HIV Directorate, Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| | - Nadia Naous
- GUM/HIV Directorate, Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| | - Lucy Garvey
- St Mary's Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Charlotte E Cohen
- GUM/HIV Directorate, Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| |
Collapse
|
207
|
Gao Z, Wu S, Yang Y, Sun M, Tian X, Jin X. Clinical characteristics of liver injury induced by immune checkpoint inhibitors in patients with advanced biliary tract carcinoma. Invest New Drugs 2023; 41:719-726. [PMID: 37589864 DOI: 10.1007/s10637-023-01391-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/02/2023] [Indexed: 08/18/2023]
Abstract
Immune-related liver injuries are closely associated with the liver's fundamental state. Patients with advanced biliary tract carcinoma (BTC) have poor liver function. We evaluated the clinical data of immune-related liver injury in patients with advanced BTC and gastric cancer (GC) during immune checkpoint inhibitor (ICI) treatment between February 2019 and July 2022 at Peking University First Hospital. Twenty-five patients with advanced BTC were identified. Fifteen patients (60%) experienced immune-related liver injury during ICI treatment. We also evaluated the clinical status of patients with GC in another group receiving immunotherapy. The results demonstrated that the incidence of immune-related liver injury was higher in patients with BTC than in GC cancer (p=0.040). Multivariate analysis suggested that the type of malignant tumor and baseline liver function status were high-risk factors for grade 2 and higher immune-related liver injuries. Two patients were diagnosed with immune-related cholangitis. Both biliary enzymes can be decreased to a certain degree by corticosteroid and ursodeoxycholic acid (UDCA) therapy but are difficult to reduce to normal levels. Liver function normalized, and symptoms improved after local treatment for cholestasis (stent implantation or PTBD). We observed a higher incidence of immune-related liver injury after ICI treatment in patients with advanced BTC. Effect of baseline liver function on the incidence of liver injury associated with immunotherapy. Interventional therapy provides rapid relief from cholestasis and is an indispensable and effective approach to the treatment of immune-related cholangitis.
Collapse
Affiliation(s)
- Zhao Gao
- Department of Medical Oncology, Peking University First Hospital, No.8, Xishiku Street, Beijing, 100034, China
| | - Shikai Wu
- Department of Medical Oncology, Peking University First Hospital, No.8, Xishiku Street, Beijing, 100034, China
| | - Yinmo Yang
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, No.8, Xishiku Street, Beijing, 100034, China
| | - Mingxia Sun
- Department of Medical Oncology, Peking University First Hospital, No.8, Xishiku Street, Beijing, 100034, China
| | - Xiaodong Tian
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, No.8, Xishiku Street, Beijing, 100034, China
| | - Xuan Jin
- Department of Medical Oncology, Peking University First Hospital, No.8, Xishiku Street, Beijing, 100034, China.
| |
Collapse
|
208
|
Banerjee P, Gaddam N, Chandler V, Chakraborty S. Oxidative Stress-Induced Liver Damage and Remodeling of the Liver Vasculature. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1400-1414. [PMID: 37355037 DOI: 10.1016/j.ajpath.2023.06.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/29/2023] [Accepted: 06/08/2023] [Indexed: 06/26/2023]
Abstract
As an organ critically important for targeting and clearing viruses, bacteria, and other foreign material, the liver operates via immune-tolerant, anti-inflammatory mechanisms indispensable to the immune response. Stress and stress-induced factors disrupt the homeostatic balance in the liver, inflicting tissue damage, injury, and remodeling. These factors include oxidative stress (OS) induced by viral infections, environmental toxins, drugs, alcohol, and diet. A recurrent theme seen among stressors common to multiple liver diseases is the induction of mitochondrial dysfunction, increased reactive oxygen species expression, and depletion of ATP. Inflammatory signaling additionally exacerbates the condition, generating a proinflammatory, immunosuppressive microenvironment and activation of apoptotic and necrotic mechanisms that disrupt the integrity of liver morphology. These pathways initiate signaling pathways that significantly contribute to the development of liver steatosis, inflammation, fibrosis, cirrhosis, and liver cancers. In addition, hypoxia and OS directly enhance angiogenesis and lymphangiogenesis in chronic liver diseases. Late-stage consequences of these conditions often narrow the outcomes for liver transplantation or result in death. This review provides a detailed perspective on various stress-induced factors and the specific focus on role of OS in different liver diseases with special emphasis on different molecular mechanisms. It also highlights how resultant changes in the liver vasculature correlate with pathogenesis.
Collapse
Affiliation(s)
- Priyanka Banerjee
- Department of Medical Physiology, Texas A&M Health Science Center, Bryan, Texas.
| | - Niyanshi Gaddam
- Department of Medical Physiology, Texas A&M Health Science Center, Bryan, Texas
| | - Vanessa Chandler
- Department of Medical Physiology, Texas A&M Health Science Center, Bryan, Texas
| | - Sanjukta Chakraborty
- Department of Medical Physiology, Texas A&M Health Science Center, Bryan, Texas.
| |
Collapse
|
209
|
Lin DSH, Tzeng SC, Cha TL, Hung CM, Lin WC, Yang CM, Lu HY, Chang JY, Huang SW. Inhalable chitosan-based hydrogel as a mucosal adjuvant for hydroxychloroquine in the treatment for SARS-CoV-2 infection in a hamster model. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2023; 56:951-960. [PMID: 37620239 DOI: 10.1016/j.jmii.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 04/20/2023] [Accepted: 08/04/2023] [Indexed: 08/26/2023]
Abstract
BACKGROUND Effective therapy for COVID-19 remains limited. Hydroxychloroquine (HCQ) has been considered, but safety and efficacy concerns remain. Chitosan exhibits antiviral and immunomodulatory effects, yet how the combination of HCQ and chitosan performs in treating COVID-19 is unknown. METHODS Male Syrian hamsters were inoculated intranasally with standardized stocks of the SARS-CoV-2 virus. Hamsters were allocated to saline (PBS), chitosan oligosaccharide (COS), HCQ, or COS + HCQ groups and received corresponding drugs. On days 1, 7, and 14 post-infection, two animals from each group were euthanized for sample collection. Viral loads were measured in lung homogenates. Biochemistry markers, cytokines, and immunoglobulins were analyzed from hamster sera. HCQ concentrations were compared between the blood, bronchoalveolar lavage, and lung tissues. All groups underwent histopathology exams of the lungs. Additional hamsters were treated with the same drugs to assess for toxicities to the heart and liver. RESULTS Among all groups, viral loads in the COS + HCQ group were the lowest by day 8. The COS + HCQ group produced the highest interleukin (IL)-6 levels on day 4, and the highest IL-10, IgA and IgG levels on day 8. HCQ concentrations were higher in the COS + HCQ group's lungs than the HCQ group, despite having received half the dose of HCQ. Histopathology demonstrated earlier inflammation resolution and swifter viral clearance in the COS + HCQ group. There was no evidence of cardiac or hepatic injury in hamsters that received HCQ. CONCLUSION In hamsters infected with the SARS-CoV-2 virus, the combination of intranasal COS and HCQ was associated with increased HCQ absorption in the lungs, more effective immune responses, without increasing the risk of hepatic or cardiac injuries.
Collapse
Affiliation(s)
- Donna Shu-Han Lin
- Division of Cardiology, Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Shian Chiuan Tzeng
- Department of Materials Science and Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan
| | - Tai-Lung Cha
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Chin-Mao Hung
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Wen-Chin Lin
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan; Department of Pathology and Graduate Institute of Pathology and Parasitology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chuen-Mi Yang
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Hsuan-Ying Lu
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Jia-Yu Chang
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Wei Huang
- Department of Orthopedics, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
210
|
Jordan VC, Sojoodi M, Shroff S, Pagan PG, Barrett SC, Wellen J, Tanabe KK, Chung RT, Caravan P, Gale EM. Molecular magnetic resonance imaging of liver inflammation using an oxidatively activated probe. JHEP Rep 2023; 5:100850. [PMID: 37818152 PMCID: PMC10561122 DOI: 10.1016/j.jhepr.2023.100850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 10/12/2023] Open
Abstract
Background & Aims Many liver diseases are driven by inflammation, but imaging to non-invasively diagnose and quantify liver inflammation has been underdeveloped. The inflammatory liver microenvironment is aberrantly oxidising owing in part to reactive oxygen species generated by myeloid leucocytes. We hypothesised that magnetic resonance imaging using the oxidatively activated probe Fe-PyC3A will provide a non-invasive biomarker of liver inflammation. Methods A mouse model of drug-induced liver injury was generated through intraperitoneal injection of a hepatoxic dose of acetaminophen. A mouse model of steatohepatitis was generated via a choline-deficient, l-amino acid defined high-fat diet (CDAHFD). Images were acquired dynamically before and after intravenous injection of Fe-PyC3A. The contrast agent gadoterate meglumine was used as a non-oxidatively activated negative control probe in mice fed CDAHFD. The (post-pre) Fe-PyC3A injection change in liver vs. muscle contrast-to-noise ratio (ΔCNR) recorded 2 min post-injection was correlated with liver function test values, histologic scoring assigned using the NASH Clinical Research Network criteria, and intrahepatic myeloid leucocyte composition determined by flow cytometry. Results For mice receiving i.p. injections of acetaminophen, intrahepatic neutrophil composition correlated poorly with liver test values but positively and significantly with ΔCNR (r = 0.64, p <0.0001). For mice fed CDAHFD, ΔCNR generated by Fe-PyC3A in the left lobe was significantly greater in mice meeting histologic criteria strongly associated with a diagnosis NASH compared to mice where histology was consistent with likely non-NASH (p = 0.0001), whereas no differential effect was observed using gadoterate meglumine. In mice fed CDAHFD, ΔCNR did not correlate strongly with fractional composition of any specific myeloid cell subpopulation as determined by flow cytometry. Conclusions Magnetic resonance imaging using Fe-PyC3A merits further evaluation as a non-invasive biomarker for liver inflammation. Impact and implications Non-invasive tests to diagnose and measure liver inflammation are underdeveloped. Inflammatory cells such as neutrophils release reactive oxygen species which creates an inflammatory liver microenvironment that can drive chemical oxidation. We recently invented a new class of magnetic resonance imaging probe that is made visible to the scanner only after chemical oxidation. Here, we demonstrate how this imaging technology could be applied as a non-invasive biomarker for liver inflammation.
Collapse
Affiliation(s)
- Veronica Clavijo Jordan
- Athinoula A. Martinos Center for Biomedical Imaging, The Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Mozhdeh Sojoodi
- Harvard Medical School, Boston, MA, USA
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Stuti Shroff
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Patricia Gonzalez Pagan
- Athinoula A. Martinos Center for Biomedical Imaging, The Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Stephen Cole Barrett
- Harvard Medical School, Boston, MA, USA
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital, Boston, MA, USA
| | | | - Kenneth K. Tanabe
- Harvard Medical School, Boston, MA, USA
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Raymond T. Chung
- Harvard Medical School, Boston, MA, USA
- Gastroenterology Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging, The Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Eric M. Gale
- Athinoula A. Martinos Center for Biomedical Imaging, The Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
211
|
Purwar S, Fatima A, Bhattacharyya H, Simhachalam Kutikuppala LV, Cozma MA, Srichawla BS, Komer L, Nurani KM, Găman MA. Toxicity of targeted anticancer treatments on the liver in myeloproliferative neoplasms. World J Hepatol 2023; 15:1021-1032. [PMID: 37900211 PMCID: PMC10600697 DOI: 10.4254/wjh.v15.i9.1021] [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] [Received: 05/28/2023] [Revised: 08/06/2023] [Accepted: 08/18/2023] [Indexed: 09/22/2023] Open
Abstract
The liver has a central role in metabolism, therefore, it is susceptible to harmful effects of ingested medications (drugs, herbs, and nutritional supplements). Drug-induced liver injury (DILI) comprises a range of unexpected reactions that occur after exposure to various classes of medication. Even though most cases consist of mild, temporary elevations in liver enzyme markers, DILI can also manifest as acute liver failure in some patients and can be associated with mortality. Herein, we briefly review available data on DILI induced by targeted anticancer agents in managing classical myeloproliferative neoplasms: Chronic myeloid leukemia, polycythemia vera, essential thrombocythemia, and myelofibrosis.
Collapse
Affiliation(s)
- Shubhrat Purwar
- Department of Internal Medicine, Grant Government Medical College, Mumbai 400008, Maharashtra, India
| | - Anam Fatima
- Department of Internal Medicine, Pandit Jawaharlal Nehru Memorial Medical College, Raipur 492001, Chhattisgarh, India
| | | | | | - Matei-Alexandru Cozma
- Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest 050474, Romania
- Department of Gastroenterology, Colentina Clinical Hospital, Bucharest 020125, Romania
| | - Bahadar Singh Srichawla
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01655, United States
| | - Leah Komer
- Department of Psychiatry, University of Toronto, Toronto M5G 1V7, Ontario, Canada
| | | | - Mihnea-Alexandru Găman
- Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest 050474, Romania
- Department of Hematology, Center of Hematology and Bone Marrow Transplantation, Fundeni Clinical Institute, Bucharest 022328, Romania.
| |
Collapse
|
212
|
Grove JI, Stephens C, Lucena MI, Andrade RJ, Weber S, Gerbes A, Bjornsson ES, Stirnimann G, Daly AK, Hackl M, Khamina-Kotisch K, Marin JJG, Monte MJ, Paciga SA, Lingaya M, Forootan SS, Goldring CEP, Poetz O, Lombaard R, Stege A, Bjorrnsson HK, Robles-Diaz M, Li D, Tran TDB, Ramaiah SK, Samodelov SL, Kullak-Ublick GA, Aithal GP. Study design for development of novel safety biomarkers of drug-induced liver injury by the translational safety biomarker pipeline (TransBioLine) consortium: a study protocol for a nested case-control study. Diagn Progn Res 2023; 7:18. [PMID: 37697410 PMCID: PMC10496294 DOI: 10.1186/s41512-023-00155-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 08/08/2023] [Indexed: 09/13/2023] Open
Abstract
A lack of biomarkers that detect drug-induced liver injury (DILI) accurately continues to hinder early- and late-stage drug development and remains a challenge in clinical practice. The Innovative Medicines Initiative's TransBioLine consortium comprising academic and industry partners is developing a prospective repository of deeply phenotyped cases and controls with biological samples during liver injury progression to facilitate biomarker discovery, evaluation, validation and qualification.In a nested case-control design, patients who meet one of these criteria, alanine transaminase (ALT) ≥ 5 × the upper limit of normal (ULN), alkaline phosphatase ≥ 2 × ULN or ALT ≥ 3 ULN with total bilirubin > 2 × ULN, are enrolled. After completed clinical investigations, Roussel Uclaf Causality Assessment and expert panel review are used to adjudicate episodes as DILI or alternative liver diseases (acute non-DILI controls). Two blood samples are taken: at recruitment and follow-up. Sample size is as follows: 300 cases of DILI and 130 acute non-DILI controls. Additional cross-sectional cohorts (1 visit) are as follows: Healthy volunteers (n = 120), controls with chronic alcohol-related or non-alcoholic fatty liver disease (n = 100 each) and patients with psoriasis or rheumatoid arthritis (n = 100, 50 treated with methotrexate) are enrolled. Candidate biomarkers prioritised for evaluation include osteopontin, glutamate dehydrogenase, cytokeratin-18 (full length and caspase cleaved), macrophage-colony-stimulating factor 1 receptor and high mobility group protein B1 as well as bile acids, sphingolipids and microRNAs. The TransBioLine project is enabling biomarker discovery and validation that could improve detection, diagnostic accuracy and prognostication of DILI in premarketing clinical trials and for clinical healthcare application.
Collapse
Affiliation(s)
- Jane I Grove
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, NG7 2UH, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - Camilla Stephens
- Servicios de Aparato Digestivo Y Farmacologia Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA Plataforma Bionand, Hospital Universitario Virgen de La Victoria, Universidad de Málaga, Malaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas Y Digestivas (CIBERehd), Madrid, Spain
| | - M Isabel Lucena
- Servicios de Aparato Digestivo Y Farmacologia Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA Plataforma Bionand, Hospital Universitario Virgen de La Victoria, Universidad de Málaga, Malaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas Y Digestivas (CIBERehd), Madrid, Spain
| | - Raúl J Andrade
- Servicios de Aparato Digestivo Y Farmacologia Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA Plataforma Bionand, Hospital Universitario Virgen de La Victoria, Universidad de Málaga, Malaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas Y Digestivas (CIBERehd), Madrid, Spain
| | - Sabine Weber
- Department of Medicine II, University Hospital, LMU Munich, Munich, Germany
| | - Alexander Gerbes
- Department of Medicine II, University Hospital, LMU Munich, Munich, Germany
| | - Einar S Bjornsson
- Department of Gastroenterology, Landspitali University Hospital Reykjavik, University of Iceland, Reykjavík, Iceland
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Guido Stirnimann
- University Clinic for Visceral Surgery and Medicine, University Hospital Inselspital and University of Bern, Bern, Switzerland
| | - Ann K Daly
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK
| | | | | | - Jose J G Marin
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas Y Digestivas (CIBERehd), Madrid, Spain
- Experimental Hepatology and Drug Targeting (HEVEPHARM), Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Maria J Monte
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas Y Digestivas (CIBERehd), Madrid, Spain
- Experimental Hepatology and Drug Targeting (HEVEPHARM), Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Sara A Paciga
- Worldwide Research Development and Medical, Pfizer, NY, USA
| | - Melanie Lingaya
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, NG7 2UH, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - Shiva S Forootan
- Centre for Drug Safety Science, University of Liverpool, Liverpool, UK
| | | | | | - Rudolf Lombaard
- ABX-CRO Advanced Pharmaceutical Services, Forschungsgesellschaft mbH, Cape Town, 7441, South Africa
| | - Alexandra Stege
- Charité-Universitätsmedizin Berlin, Central Biobank Charité (ZeBanC), Berlin, Germany
| | - Helgi K Bjorrnsson
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mercedes Robles-Diaz
- Servicios de Aparato Digestivo Y Farmacologia Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA Plataforma Bionand, Hospital Universitario Virgen de La Victoria, Universidad de Málaga, Malaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas Y Digestivas (CIBERehd), Madrid, Spain
| | - Dingzhou Li
- Worldwide Research Development and Medical, Pfizer, NY, USA
| | | | | | - Sophia L Samodelov
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, 8006, Zurich, Switzerland
| | - Gerd A Kullak-Ublick
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, 8006, Zurich, Switzerland
- Mechanistic Safety, CMO & Patient Safety, Global Drug Development, Novartis Pharma, 4056, Basel, Switzerland
| | - Guruprasad P Aithal
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, NG7 2UH, UK.
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK.
| |
Collapse
|
213
|
Phansalkar R, Kambham N, Charu V. Liver Pathology After Hematopoietic Stem Cell Transplantation. Surg Pathol Clin 2023; 16:519-532. [PMID: 37536886 DOI: 10.1016/j.path.2023.04.007] [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] [Indexed: 08/05/2023]
Abstract
Hematopoietic stem cell transplantation is used to treat a variety of hematologic malignancies and autoimmune conditions. The immunosuppressive medications as well as other therapies used both before and after transplantation leave patients susceptible to a wide spectrum of complications, including liver injury. Causes for liver damage associated with stem cell transplantation include sinusoidal obstruction syndrome, graft-versus-host disease, iron overload, and opportunistic infection. Here, the authors review the clinical and pathological findings of these etiologies of liver injury and provide a framework for diagnosis.
Collapse
Affiliation(s)
- Ragini Phansalkar
- Department of Pathology, Lane Building, L235, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Neeraja Kambham
- Department of Pathology, Lane Building, L235, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Vivek Charu
- Department of Pathology, Lane Building, L235, 300 Pasteur Drive, Stanford, CA 94305, USA; Department of Medicine, Quantitative Sciences Unit, Stanford, CA, USA.
| |
Collapse
|
214
|
Williamson C, Nana M, Poon L, Kupcinskas L, Painter R, Taliani G, Heneghan M, Marschall HU, Beuers U. EASL Clinical Practice Guidelines on the management of liver diseases in pregnancy. J Hepatol 2023; 79:768-828. [PMID: 37394016 DOI: 10.1016/j.jhep.2023.03.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 03/10/2023] [Indexed: 07/04/2023]
Abstract
Liver diseases in pregnancy comprise both gestational liver disorders and acute and chronic hepatic disorders occurring coincidentally in pregnancy. Whether related to pregnancy or pre-existing, liver diseases in pregnancy are associated with a significant risk of maternal and fetal morbidity and mortality. Thus, the European Association for the Study of Liver Disease invited a panel of experts to develop clinical practice guidelines aimed at providing recommendations, based on the best available evidence, for the management of liver disease in pregnancy for hepatologists, gastroenterologists, obstetric physicians, general physicians, obstetricians, specialists in training and other healthcare professionals who provide care for this patient population.
Collapse
|
215
|
Andújar-Vera F, Alés-Palmer ML, Muñoz-de-Rueda P, Iglesias-Baena I, Ocete-Hita E. Metabolomic Analysis of Pediatric Patients with Idiosyncratic Drug-Induced Liver Injury According to the Updated RUCAM. Int J Mol Sci 2023; 24:13562. [PMID: 37686369 PMCID: PMC10487599 DOI: 10.3390/ijms241713562] [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: 07/28/2023] [Revised: 08/20/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
Hepatotoxicity, a common adverse drug effect, has been extensively studied in adult patients. However, it is equally important to investigate this condition in pediatric patients to develop personalized treatment strategies for children. This study aimed to identify plasma biomarkers that characterize hepatotoxicity in pediatric patients through an observational case-control study. Metabolomic analysis was conducted on 55 pediatric patients with xenobiotic liver toxicity and 88 healthy controls. The results revealed clear differences between the two groups. Several metabolites, including hydroxydecanoylcarnitine, octanoylcarnitine, lysophosphatidylcholine, glycocholic acid, and taurocholic acid, were identified as potential biomarkers (area under the curve: 0.817; 95% confidence interval: 0.696-0.913). Pathway analysis indicated involvement of primary bile acid biosynthesis and the metabolism of taurine and hypotaurine (p < 0.05). The findings from untargeted metabolomic analysis demonstrated an increase in bile acids in children with hepatotoxicity. The accumulation of cytotoxic bile acids should be further investigated to elucidate the role of these metabolites in drug-induced liver injury.
Collapse
Affiliation(s)
| | - María Luisa Alés-Palmer
- Department of Pediatrics, University of Granada, 18016 Granada, Spain;
- Department of Pediatrics, “Virgen de las Nieves” University Hospital, 18014 Granada, Spain
| | - Paloma Muñoz-de-Rueda
- Research Support Unit, Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain;
| | | | - Esther Ocete-Hita
- Department of Pediatrics, University of Granada, 18016 Granada, Spain;
- Department of Pediatrics, “Virgen de las Nieves” University Hospital, 18014 Granada, Spain
- Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
| |
Collapse
|
216
|
Andrade RJ, Aithal GP, de Boer YS, Liberal R, Gerbes A, Regev A, Terziroli Beretta-Piccoli B, Schramm C, Kleiner DE, De Martin E, Kullak-Ublick GA, Stirnimann G, Devarbhavi H, Vierling JM, Manns MP, Sebode M, Londoño MC, Avigan M, Robles-Diaz M, García-Cortes M, Atallah E, Heneghan M, Chalasani N, Trivedi PJ, Hayashi PH, Taubert R, Fontana RJ, Weber S, Oo YH, Zen Y, Licata A, Lucena MI, Mieli-Vergani G, Vergani D, Björnsson ES. Nomenclature, diagnosis and management of drug-induced autoimmune-like hepatitis (DI-ALH): An expert opinion meeting report. J Hepatol 2023; 79:853-866. [PMID: 37164270 PMCID: PMC10735171 DOI: 10.1016/j.jhep.2023.04.033] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 04/26/2023] [Accepted: 04/29/2023] [Indexed: 05/12/2023]
Abstract
Drug-induced liver injury (DILI) can mimic almost all other liver disorders. A phenotype increasingly ascribed to drugs is autoimmune-like hepatitis (ALH). This article summarises the major topics discussed at a joint International Conference held between the Drug-Induced Liver Injury consortium and the International Autoimmune Hepatitis Group. DI-ALH is a liver injury with laboratory and/or histological features that may be indistinguishable from those of autoimmune hepatitis (AIH). Previous studies have revealed that patients with DI-ALH and those with idiopathic AIH have very similar clinical, biochemical, immunological and histological features. Differentiating DI-ALH from AIH is important as patients with DI-ALH rarely require long-term immunosuppression and the condition often resolves spontaneously after withdrawal of the implicated drug, whereas patients with AIH mostly require long-term immunosuppression. Therefore, revision of the diagnosis on long-term follow-up may be necessary in some cases. More than 40 different drugs including nitrofurantoin, methyldopa, hydralazine, minocycline, infliximab, herbal and dietary supplements (such as Khat and Tinospora cordifolia) have been implicated in DI-ALH. Understanding of DI-ALH is limited by the lack of specific markers of the disease that could allow for a precise diagnosis, while there is similarly no single feature which is diagnostic of AIH. We propose a management algorithm for patients with liver injury and an autoimmune phenotype. There is an urgent need to prospectively evaluate patients with DI-ALH systematically to enable definitive characterisation of this condition.
Collapse
Affiliation(s)
- Raúl J Andrade
- Servicio Aparato Digestivo and Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA_Plataforma Bionand, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain.
| | - Guruprasad P Aithal
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - Ynto S de Boer
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, VU University Medical Center, Amsterdam, Netherlands
| | - Rodrigo Liberal
- Gastroenterology Department, Centro Hospitalar Universitário de São João, Porto, Portugal; Faculty of Medicine of the University of Porto, Porto, Portugal
| | | | - Arie Regev
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Christoph Schramm
- Department of Medicine, University Medical Center Hamburg-Eppendorf. Hamburg Center for Translational Immunology. Martin Zeitz Center for Rare Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - David E Kleiner
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, USA
| | - Eleonora De Martin
- APHP, Hôpital Paul Brousse, Centre Hépato-Biliaire, INSERM Unit 1193, FHU Hepatinov, Villejuif, France
| | - Gerd A Kullak-Ublick
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Mechanistic Safety, Global Drug Development, Novartis, Basel, Switzerland
| | - Guido Stirnimann
- Department of Visceral Surgery and Medicine, Inselspital University Hospital and University of Bern, Bern, Switzerland
| | - Harshad Devarbhavi
- Department of Gastroenterology and Hepatology, St. John's Medical College Hospital, Bangalore, India
| | - John M Vierling
- Departments of Medicine and Surgery, Section of Gastroenterology and Hepatology and Division of Abdominal Transplantation, Baylor College of Medicine, Houston, Texas, United States
| | - Michael P Manns
- Hannover Medical School, Centre of ERN RARE-LIVER, Hannover, Germany
| | - Marcial Sebode
- Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Hamburg, Germany
| | - Maria Carlota Londoño
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain; Liver Unit, Hospital Clínic de Barcelona, Health Care Provider of the European Reference Network on Rare Liver Disorders (ERN-Liver), Institut d' Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Mark Avigan
- Center for Drug Evaluation and Research, Office of Surveillance and Epidemiology, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Mercedes Robles-Diaz
- Servicio Aparato Digestivo and Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA_Plataforma Bionand, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Miren García-Cortes
- Servicio Aparato Digestivo and Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA_Plataforma Bionand, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Edmond Atallah
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | | | - Naga Chalasani
- University School of Medicine & Indiana University Health, Indianapolis, Indiana, USA
| | - Palak J Trivedi
- NIHR Birmingham BRC, Institute of Immunology and Immunotherapy, Centre for Liver and Gastrointestinal Research, Liver Unit, University Hospitals Birmingham National Health Service Foundation Trust Queen Elizabeth, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Paul H Hayashi
- Division of Hepatology and Nutrition, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Richard Taubert
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Hannover, Germany
| | - Robert J Fontana
- Division of Gastroenterology and Hepatology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Sabine Weber
- Department of Medicine II, LMU Klinikum Munich, Munich, Germany
| | - Ye Htun Oo
- Center for Liver and Gastro Research & National Institute of Health Research Birmingham Biomedical Research Centre, University of Birmingham, Centre for Rare Disease and ERN Rare Liver Centre, Liver Transplant and Hepatobiliary Unit, University Hospital Birmingham NHS Foundation Trust, UK
| | - Yoh Zen
- Institute of Liver Studies, King's College Hospital, London SE5 9RS, UK
| | - Anna Licata
- Medicina Interna ed Epatologia, Università degli Studi di Palermo, Palermo, Italy
| | - M Isabel Lucena
- Servicio Aparato Digestivo and Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA_Plataforma Bionand, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain; Platform ISCiii for Clinical Research and Clinical Trials SCReN UICEC- IBIMA, Málaga, Spain.
| | - Giorgina Mieli-Vergani
- MowatLabs, Faculty of Life Sciences and Medicine, King's College London, King's College Hospital, London, United Kingdom
| | - Diego Vergani
- MowatLabs, Faculty of Life Sciences and Medicine, King's College London, King's College Hospital, London, United Kingdom
| | - Einar S Björnsson
- Faculty of Medicine, University of Iceland, Department of Gastroenterology and Hepatology, Landspitali University Hospital, Reykjavik, Iceland
| |
Collapse
|
217
|
Du X, Liu Z, Yu H, Wang Y, Zou Z, Wei H, Liang J, Yang D, Liu Y, Zhang J, Pan CQ. Prognostic risk factors for patients with hepatic sinusoidal obstruction syndrome caused by pyrrolizidine alkaloids. Medicine (Baltimore) 2023; 102:e34698. [PMID: 37565875 PMCID: PMC10419670 DOI: 10.1097/md.0000000000034698] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 07/20/2023] [Indexed: 08/12/2023] Open
Abstract
Pyrrolizidine alkaloids induced hepatic sinusoidal obstruction syndrome (PA-HSOS) often occurs after consuming herbs or a dietary supplement containing the plant Tu-San-Qi. Limited data exists to identify patients with fatal outcomes for early interventions. We aimed to analyze the predictors for 3-month survival. We retrospectively enrolled PA-HSOS patients in 5 hospitals and extracted data from the onset of PA-HSOS to 36 months. Outcome measurements were 3-month and 36-month survival rates, baseline prognostic predictors for survival, and the effects of anticoagulant therapy. Among 49 enrollees, the median age was 60 and 49% male. At the onset of PA-HSOS, patients with Child-Turcotte-Pugh (CTP) class of A, B, or C were 8.2% (4/49), 42.8% (21/49) and 49.0% (24/49), respectively. None of them received a transjugular intrahepatic portosystemic shunt or a liver transplant. The 3-month and 36-month survival rates were 86% and 76%, respectively. Compared to the CTP class A or B, class C at baseline independently predicted lower survival rates at both 3 and 36 months. However, anticoagulation therapy treatment within the first 3 months independently predicted significantly higher survival rates at both time points. CTP class C and anticoagulant therapy were the independent predictors for short-term and long-term survival. Anticoagulant therapy could decrease mortality rate of CTP class C patients. The greatest benefit of anticoagulant evaluated by 3-month survival rate was in patients with CTP class C compared with those without treatment (93% vs 40%, P = .009). There were no bleeding complications reported in patients treated with the anticoagulant.
Collapse
Affiliation(s)
- Xiaofei Du
- Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Zhenli Liu
- Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Haibin Yu
- Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yu Wang
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Zhengsheng Zou
- The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Hongshan Wei
- Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Jing Liang
- Tianjin Third Central Hospital, Tianjin, China
| | - Daokun Yang
- The First Affiliated Hospital of Xinxiang Medical University, Henan, China
| | - Yali Liu
- Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Jing Zhang
- Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Calvin Q. Pan
- Division on Gastroenterology and Hepatology, NYU Langone Health, NYU Grossman School of Medicine, NY
| |
Collapse
|
218
|
Li X, Ni J, Chen L. Advances in the study of acetaminophen-induced liver injury. Front Pharmacol 2023; 14:1239395. [PMID: 37601069 PMCID: PMC10436315 DOI: 10.3389/fphar.2023.1239395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 07/28/2023] [Indexed: 08/22/2023] Open
Abstract
Acetaminophen (APAP) overdose is a significant cause of drug-induced liver injury and acute liver failure. The diagnosis, screening, and management of APAP-induced liver injury (AILI) is challenging because of the complex mechanisms involved. Starting from the current studies on the mechanisms of AILI, this review focuses on novel findings in the field of diagnosis, screening, and management of AILI. It highlights the current issues that need to be addressed. This review is supposed to summarize the recent research progress and make recommendations for future research.
Collapse
Affiliation(s)
- Xinghui Li
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Jiaqi Ni
- West China School of Pharmacy, Sichuan University, Chengdu, China
- Department of Pharmacy, Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Li Chen
- Department of Pharmacy, Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| |
Collapse
|
219
|
Barouki R, Samson M, Blanc EB, Colombo M, Zucman-Rossi J, Lazaridis KN, Miller GW, Coumoul X. The exposome and liver disease - how environmental factors affect liver health. J Hepatol 2023; 79:492-505. [PMID: 36889360 PMCID: PMC10448911 DOI: 10.1016/j.jhep.2023.02.034] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 03/10/2023]
Abstract
Since the initial development of the exposome concept, much effort has been devoted to the characterisation of the exposome through analytical, epidemiological, and toxicological/mechanistic studies. There is now an urgent need to link the exposome to human diseases and to include exposomics in the characterisation of environment-linked pathologies together with genomics and other omics. Liver diseases are particularly well suited for such studies since major functions of the liver include the detection, detoxification, and elimination of xenobiotics, as well as inflammatory responses. It is well known that several liver diseases are associated with i) addictive behaviours such as alcohol consumption, smoking, and to a certain extent dietary imbalance and obesity, ii) viral and parasitic infections, and iii) exposure to toxins and occupational chemicals. Recent studies indicate that environmental exposures are also significantly associated with liver diseases, and these include air pollution (particulate matter and volatile chemicals), contaminants such as polyaromatic hydrocarbons, bisphenol A and per-and poly-fluorinated substances, and physical stressors such as radiation. Furthermore, microbial metabolites and the "gut-liver" axis play a major role in liver diseases. Exposomics is poised to play a major role in the field of liver pathology. Methodological advances such as the exposomics-metabolomics framework, the determination of risk factors' genomic and epigenomic signatures, and cross-species biological pathway analysis should further delineate the impact of the exposome on the liver, opening the way for improved prevention, as well as the identification of new biomarkers of exposure and effects, and additional therapeutic targets.
Collapse
Affiliation(s)
| | - Michel Samson
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | | | - Massimo Colombo
- San Raffaele Hospital, Liver Center, Via Olgettina 60, 20132, Milan, Italy
| | - Jessica Zucman-Rossi
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Inserm, AP-HP, Hôpital Européen Georges Pompidou, Institut du Cancer Paris CARPEM, F-75006, Paris, France
| | | | - Gary W Miller
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, 10032, USA
| | | |
Collapse
|
220
|
Ma J, Ghabril M, Chalasani N. Drug-Induced Acute-on-Chronic Liver Failure: Challenges and Future Directions. Clin Liver Dis 2023; 27:631-648. [PMID: 37380287 DOI: 10.1016/j.cld.2023.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
Drug-induced liver injury (DILI) is a global problem related to prescription and over-the-counter medications as well as herbal and dietary supplements. It can lead to liver failure with the risk of death and need for liver transplantation. Acute-on-chronic liver failure (ACLF) may be precipitated by DILI and is associated with a high risk of mortality. This review addresses the challenges in defining the diagnostic criteria of drug-induced ACLF (DI-ACLF). The studies characterizing DI-ACLF and its outcomes are summarized, highlighting geographic differences in underlying liver disease and implicated agents, as are future directions in the field.
Collapse
Affiliation(s)
- Jiayi Ma
- Gastroenterology and Hepatology, Indiana University School of Medicine, 702 Rotary Circle, Suite 225, Indianapolis, IN 46202, USA
| | - Marwan Ghabril
- Gastroenterology and Hepatology, Indiana University School of Medicine & Indiana University Health, 702 Rotary Circle, Suite 225, Indianapolis, IN 46202, USA
| | - Naga Chalasani
- Gastroenterology and Hepatology, Indiana University School of Medicine & Indiana University Health, 702 Rotary Circle, Suite 225, Indianapolis, IN 46202, USA.
| |
Collapse
|
221
|
Lim J, Kim JS, Kim HW, Kim YH, Jung SS, Kim JW, Oh JY, Lee H, Kim SK, Kim SH, Lyu J, Ko Y, Kwon SJ, Jeong YJ, Kim DJ, Koo HK, Jegal Y, Kyung SY, An TJ, Min J. Metabolic Disorders Are Associated With Drug-Induced Liver Injury During Antituberculosis Treatment: A Multicenter Prospective Observational Cohort Study in Korea. Open Forum Infect Dis 2023; 10:ofad422. [PMID: 37654787 PMCID: PMC10468151 DOI: 10.1093/ofid/ofad422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/07/2023] [Indexed: 09/02/2023] Open
Abstract
Background Drug-induced liver injury (DILI) may lead to the discontinuation of antituberculosis (anti-TB) treatment (ATT). Some studies have suggested that metabolic disorders increase the risk of DILI during ATT. This study aimed to identify risk factors for DILI, particularly metabolic disorders, during ATT. Methods A multicenter prospective observational cohort study to evaluate adverse events during ATT was conducted in Korea from 2019 to 2021. Drug-susceptible patients with TB who had been treated with standard ATT for 6 months were included. The patients were divided into 2 groups depending on the presence of 1 or more metabolic conditions, such as insulin resistance, hypertension, obesity, and dyslipidemia. We monitored ATT-related adverse events, including DILI, and treatment outcomes. The incidence of DILI was compared between individuals with and without metabolic disorders, and related factors were evaluated. Results Of 684 patients, 52 (7.6%) experienced DILI, and 92.9% of them had metabolic disorders. In the multivariable analyses, underlying metabolic disorders (adjusted hazard ratio [aHR], 2.85; 95% CI, 1.01-8.07) and serum albumin <3.5 g/dL (aHR, 2.26; 95% CI, 1.29-3.96) were risk factors for DILI during ATT. In the 1-month landmark analyses, metabolic disorders were linked to an elevated risk of DILI, especially significant alanine aminotransferase elevation. The treatment outcome was not affected by the presence of metabolic disorders. Conclusions Patients with metabolic disorders have an increased risk of ATT-induced liver injury compared with controls. The presence of metabolic disorders and hypoalbuminemia adversely affects the liver in patients with ATT.
Collapse
Affiliation(s)
- Jihye Lim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ju Sang Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyung Woo Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yong Hyun Kim
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sung Soo Jung
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Jin Woo Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jee Youn Oh
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Heayon Lee
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sung Kyoung Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sun-Hyung Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Republic of Korea
| | - Jiwon Lyu
- Department of Pulmonary and Critical Care Medicine, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine, Cheonan, Republic of Korea
| | - Yousang Ko
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Sun Jung Kwon
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Konyang University Hospital, Konyang University College of Medicine, Daejeon, Republic of Korea
| | - Yun-Jeong Jeong
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Dongguk University Ilsan Hospital, Goyang, Republic of Korea
| | - Do Jin Kim
- Division of Allergy and Respiratory Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Republic of Korea
| | - Hyeon-Kyoung Koo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Republic of Korea
| | - Yangjin Jegal
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Ulsan University Hospital, Ulsan University College of Medicine, Ulsan, Republic of Korea
| | - Sun Young Kyung
- Division of Pulmonology, Departments of Internal Medicine, Gachon University Gil Hospital, Incheon, Republic of Korea
| | - Tai Joon An
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jinsoo Min
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
222
|
Wang Y, Ma J, Ma S, Wang J, Li J. Causal Evaluation of Post-Marketing Drugs for Drug-induced Liver Injury from Electronic Health Records. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2023; 2023:1-4. [PMID: 38083643 DOI: 10.1109/embc40787.2023.10340721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Drug-induced liver injury (DILI) is one of the most common and serious adverse drug reactions that can lead to acute liver failure and death. Detection of DILI and causal estimation of drug-hepatotoxicity association are of great importance for patient safety. This paper proposes a framework for causal estimation of post-marketing drugs for DILI from real-world electronic health record (EHR) data. Randomized clinical trials were replicated at scale by automatically generating different user and non-user cohorts for each potential drug, and average treatment effects (ATEs) of drugs were estimated using targeted maximum likelihood estimation. Ten years of real-world EHRs were used to validate the framework. Of all 1199 single-ingredient drugs analyzed, 7 novel and 7 known drug-hepatotoxicity associations were found to be causal.
Collapse
|
223
|
Guirguis N, Bertrand A, Rose CF, Matoori S. 175 Years of Bilirubin Testing: Ready for Point-of-Care? Adv Healthc Mater 2023; 12:e2203380. [PMID: 37035945 PMCID: PMC11468846 DOI: 10.1002/adhm.202203380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/26/2023] [Indexed: 04/11/2023]
Abstract
Bilirubin was first detected in blood in 1847 and since then has become one of the most widely used biomarkers for liver disease. Clinical routine bilirubin testing is performed at the hospital laboratory, and the gold standard colorimetric test is prone to interferences. The absence of a bedside test for bilirubin delays critical clinical decisions for patients with liver disease. This clinical care gap has motivated the development of a new generation of bioengineered point-of-care bilirubin assays. In this Perspective, recently developed bilirubin assays are critically discussed, and their translational potential evaluated.
Collapse
Affiliation(s)
- Natalie Guirguis
- Faculté de PharmacieUniversité de MontréalMontrealQCH3T 1J4Canada
| | | | - Christopher F. Rose
- Hepato‐Neuro LaboratoryCentre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQCH2X 0A9Canada
- Department of MedicineUniversité de MontréalMontrealQCH3T 1J4Canada
| | - Simon Matoori
- Faculté de PharmacieUniversité de MontréalMontrealQCH3T 1J4Canada
| |
Collapse
|
224
|
Fontana RJ, Bjornsson ES, Reddy R, Andrade RJ. The Evolving Profile of Idiosyncratic Drug-Induced Liver Injury. Clin Gastroenterol Hepatol 2023; 21:2088-2099. [PMID: 36868489 DOI: 10.1016/j.cgh.2022.12.040] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/02/2022] [Accepted: 12/19/2022] [Indexed: 03/05/2023]
Abstract
Idiosyncratic drug-induced liver injury (DILI) is an infrequent but important cause of liver disease. Newly identified causes of DILI include the COVID vaccines, turmeric, green tea extract, and immune checkpoint inhibitors. DILI is largely a clinical diagnosis of exclusion that requires evaluation for more common causes of liver injury and a compatible temporal association with the suspect drug. Recent progress in DILI causality assessment includes the development of the semi-automated revised electronic causality assessment method (RECAM) instrument. In addition, several drug-specific HLA associations have been identified that can help with the confirmation or exclusion of DILI in individual patients. Various prognostic models can help identify the 5%-10% of patients at highest risk of death. Following suspect drug cessation, 80% of patients with DILI fully recover, whereas 10%-15% have persistently abnormal laboratory studies at 6 months of follow-up. Hospitalized patients with DILI with an elevated international normalized ratio or mental status changes should be considered for N-acetylcysteine therapy and urgent liver transplant evaluation. Selected patients with moderate to severe drug reaction with eosinophilia and systemic symptoms or autoimmune features on liver biopsy may benefit from short-term corticosteroids. However, prospective studies are needed to determine the optimal patients and dose and duration of steroids to use. LiverTox is a comprehensive, freely accessible Web site with important information regarding the hepatotoxicity profile of more than 1000 approved medications and 60 herbal and dietary supplement products. It is hoped that ongoing "omics" studies will lead to additional insight into DILI pathogenesis, improved diagnostic and prognostic biomarkers, and mechanism-based treatments.
Collapse
Affiliation(s)
- Robert J Fontana
- Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, Michigan.
| | - Einar S Bjornsson
- Deparment of Internal Medicine, Landspitali University Hospital, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Rajender Reddy
- Division of Gastroenterology and Hepatology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Raul J Andrade
- Division of Gastroenterology and Hepatology, University Hospital-IBIMA Platform BIONAND, University of Malaga, CIBERehd, Spain
| |
Collapse
|
225
|
Roeb E. [Drug toxic hepatopathy - an underestimated danger]. Dtsch Med Wochenschr 2023; 148:828-835. [PMID: 37364577 DOI: 10.1055/a-1871-6426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Drug-induced toxic hepatopathies and drug-induced liver injury, DILI, are characterized by a variety of clinical manifestations and therefore represent a significant diagnostic challenge. This article shows how DILI is diagnosed and what therapy options exist. Current special cases of DILI genesis are also discussed (DOACs, IBD drugs, tyrosine kinase inhibitors). These newer substances and corresponding hepatotoxic effects are not yet fully understood. The internationally recognized and online available RUCAM score (Roussel Uclaf Causality Assessment Method) helps to assess the probability of drug-related toxic liver damage.
Collapse
|
226
|
Dong Z, Si G, Zhu X, Li C, Hua R, Teng J, Zhang W, Xu L, Qian W, Liu B, Wang J, Wang T, Tang Y, Zhao Y, Gong X, Tao Z, Xu Z, Li Y, Chen B, Kong X, Xu Y, Gu N, Li C. Diagnostic Performance and Safety of a Novel Ferumoxytol-Enhanced Coronary Magnetic Resonance Angiography. Circ Cardiovasc Imaging 2023; 16:580-590. [PMID: 37463240 DOI: 10.1161/circimaging.123.015404] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 06/13/2023] [Indexed: 07/20/2023]
Abstract
BACKGROUND Currently, noninvasive arteriography for the diagnosis of coronary artery disease is clinically limited to the computed tomography scanning, where patients have to be exposed to the radiation and risks associated with iodinated contrast. We aimed to investigate the diagnostic performance and safety of a novel ferumoxytol-enhanced coronary magnetic resonance angiography (CMRA) in patients with suspected coronary artery disease. METHODS Thirty patients, 19 males, with a median age of 63 years old, and 17 with renal insufficiency, who were scheduled for invasive coronary angiography, were enrolled. Ferumoxytol was administered intravenously with a dose of 3 mg/kg during CMRA. Images were acquired with an ECG-triggered, navigator-gated, inversion recovery-prepared 3D fast low-angle shot sequence, and the image quality was assessed by a 4-point scale. Eighteen-segment coronary artery model was adopted to evaluate the visibility of the coronary arteries, and the image quality and stenosis were evaluated in nine segments. The diagnostic performance of CMRA is described as sensitivity, specificity, positive and negative predictive values, and accuracy with the invasive coronary angiography results as reference. The patients' vital signs were monitored during CMRA, and their hepatic and renal functions were followed up for 3 months to evaluate the safety of ferumoxytol. RESULTS Two hundred fifty-two of the 270 study segments were identified by CMRA, and their quality score reached 3.6±0.7. Referring to the invasive coronary angiography results, the sensitivity, specificity, positive predictive value, negative predictive value, and accuracy of ferumoxytol-enhanced CMRA reached 100.0%, 66.7%, 92.3%, 100.0%, and 93.3% respectively in patient-based analysis; 91.4%, 90.9%, 86.5%, 94.3%, and 91.1%, respectively in vessel-based analysis; and 92.3%, 96.7%, 83.7%, 98.6%, and 96.0%, respectively in segment-based analysis. No ferumoxytol-related adverse event was observed during the 3-month follow-up. CONCLUSIONS Ferumoxytol-enhanced CMRA demonstrated good diagnostic performance and excellent safety in the diagnosis of significant coronary stenosis, providing an alternative to coronary computed tomography angiography for the diagnosis of coronary artery disease. REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT05032937.
Collapse
Affiliation(s)
- Zhou Dong
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (Z.D., C.L., R.H., J.T., W.Z., T.W., X.G., Z.T., Z.X., Y.L., B.C., X.K., C.L.)
| | - Guangxiang Si
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, China (G.S., N.G.)
| | - Xiaomei Zhu
- Department of Radiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (X.Z., L.X., W.Q., B.L., J.W., Y.X.)
| | - Chen Li
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (Z.D., C.L., R.H., J.T., W.Z., T.W., X.G., Z.T., Z.X., Y.L., B.C., X.K., C.L.)
| | - Rui Hua
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (Z.D., C.L., R.H., J.T., W.Z., T.W., X.G., Z.T., Z.X., Y.L., B.C., X.K., C.L.)
| | - Jianzhen Teng
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (Z.D., C.L., R.H., J.T., W.Z., T.W., X.G., Z.T., Z.X., Y.L., B.C., X.K., C.L.)
| | - Wenhao Zhang
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (Z.D., C.L., R.H., J.T., W.Z., T.W., X.G., Z.T., Z.X., Y.L., B.C., X.K., C.L.)
| | - Lulu Xu
- Department of Radiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (X.Z., L.X., W.Q., B.L., J.W., Y.X.)
| | - Wen Qian
- Department of Radiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (X.Z., L.X., W.Q., B.L., J.W., Y.X.)
| | - Bo Liu
- Department of Radiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (X.Z., L.X., W.Q., B.L., J.W., Y.X.)
| | - Jun Wang
- Department of Radiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (X.Z., L.X., W.Q., B.L., J.W., Y.X.)
| | - Tong Wang
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (Z.D., C.L., R.H., J.T., W.Z., T.W., X.G., Z.T., Z.X., Y.L., B.C., X.K., C.L.)
| | - Yingdan Tang
- Department of Biostatistics, School of Public Health, Nanjing Medical University, No. 101 Longmian Avenue, Nanjing, China (Y.T., Y.Z.)
| | - Yang Zhao
- Department of Biostatistics, School of Public Health, Nanjing Medical University, No. 101 Longmian Avenue, Nanjing, China (Y.T., Y.Z.)
| | - Xiaoxuan Gong
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (Z.D., C.L., R.H., J.T., W.Z., T.W., X.G., Z.T., Z.X., Y.L., B.C., X.K., C.L.)
| | - Zhiwen Tao
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (Z.D., C.L., R.H., J.T., W.Z., T.W., X.G., Z.T., Z.X., Y.L., B.C., X.K., C.L.)
| | - Zhihui Xu
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (Z.D., C.L., R.H., J.T., W.Z., T.W., X.G., Z.T., Z.X., Y.L., B.C., X.K., C.L.)
| | - Yong Li
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (Z.D., C.L., R.H., J.T., W.Z., T.W., X.G., Z.T., Z.X., Y.L., B.C., X.K., C.L.)
| | - Bo Chen
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (Z.D., C.L., R.H., J.T., W.Z., T.W., X.G., Z.T., Z.X., Y.L., B.C., X.K., C.L.)
| | - Xiangqing Kong
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (Z.D., C.L., R.H., J.T., W.Z., T.W., X.G., Z.T., Z.X., Y.L., B.C., X.K., C.L.)
| | - Yi Xu
- Department of Radiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (X.Z., L.X., W.Q., B.L., J.W., Y.X.)
| | - Ning Gu
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (Z.D., C.L., R.H., J.T., W.Z., T.W., X.G., Z.T., Z.X., Y.L., B.C., X.K., C.L.)
- Medical School, Nanjing University, Nanjing, Jiangsu, China (N.G.)
| | - Chunjian Li
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (Z.D., C.L., R.H., J.T., W.Z., T.W., X.G., Z.T., Z.X., Y.L., B.C., X.K., C.L.)
| |
Collapse
|
227
|
Sukumaran D, Usharani P, Paramjyothi GK, Subbalaxmi MVS, Sireesha K, Abid Ali M. A study to evaluate the hepatoprotective effect of N- acetylcysteine on anti tuberculosis drug induced hepatotoxicity and quality of life. Indian J Tuberc 2023; 70:303-310. [PMID: 37562904 DOI: 10.1016/j.ijtb.2022.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 05/26/2022] [Indexed: 08/12/2023]
Abstract
BACKGROUND Drug induced liver injury (DILI) is a serious adverse effect caused by first-line anti-TB (ATT) drugs, limiting the TB-treatment. The tissue inflammation induced by free radical burst and poor dietary intake in TB induces oxidative stress, which was proposed as one of the mechanisms responsible for ATT induced DILI. N-acetylcysteine (NAC) exerts a hepato-protective effect by enhancing the cellular antioxidant defense mechanism. There are few studies evaluating the effect of NAC on ATT induced DILI in Indian-population. METHODS This is a prospective, randomized, double-blind, placebo-controlled, parallel-group study. Thirty-eight newly diagnosed TB patients on first-line ATT with normal liver function test (LFT) were recruited and randomized to receive either NAC 600 mg tablet or placebo twice daily for 4 weeks and followed-up for next 4 weeks. LFT [AST, ALT, ALP and Total bilirubin] was assessed at baseline, 2, 4 and 8 weeks. Oxidative-stress biomarkers [Malondialdehyde (MDA), Nitric Oxide (NO), Glutathione (GSH)] and quality of life (QOL) by SF-36 questionnaire were assessed at baseline, 4 and 8 weeks. Adverse Drug Reactions (ADRs) were monitored at every visit. Compliance was assessed by pill-count method. RESULTS Baseline characteristics were homogenous among both the groups. In the NAC group, there was significant reduction in ALT (p < 0.01), ALP (p < 0.01), total bilirubin (p < 0.001) at 4 weeks compared to baseline. AST, MDA and NO showed a reduction of 19%, 21.6% and 5.5% respectively from baseline and GSH at showed an increase of 2.6% from baseline at 4 weeks in the NAC group, however these were not statistically significant. These effects in LFT and oxidative biomarkers persisted even at the end of 8 weeks. Significant improvement from baseline in QOL was observed in both the groups (p < 0.05). Between group analysis showed, significant reduction in ALT (p < 0.05) and AST (p < 0.05) in NAC group at 4 weeks, whereas bilirubin, MDA, NO and GSH showed improvement at 4 weeks compared to placebo in NAC group, however it was not statistically significant. This improvement in the LFT and oxidative biomarkers continued even at the end of 8 weeks. Itching and rashes were the most common ADRs, with similar incidence in both the groups. Compliance to treatment was good in both the groups. CONCLUSION Significant improvement in liver function parameters is suggestive of hepatoprotective effect of NAC. This observed effect at 4 weeks was found to be persistent at 8 weeks, which signifies prolonged hepato-protective effect of NAC. Long duration studies with large sample size are required for further confirmation of hepato-protective action of NAC.
Collapse
Affiliation(s)
- Deepasree Sukumaran
- Department of Clinical Pharmacology and Therapeutics, Nizam's Institute of Medical Sciences, Hyderabad.
| | - P Usharani
- Department of Clinical Pharmacology and Therapeutics, Nizam's Institute of Medical Sciences, Hyderabad
| | - G K Paramjyothi
- Department of Pulmonary Medicine, Nizam's Institute of Medical Sciences, Hyderabad
| | - M V S Subbalaxmi
- Department of General Medicine, Nizam's Institute of Medical Sciences, Hyderabad
| | - K Sireesha
- Department of Clinical Pharmacology and Therapeutics, Nizam's Institute of Medical Sciences, Hyderabad
| | - Mohammed Abid Ali
- Department of Clinical Pharmacology and Therapeutics, Nizam's Institute of Medical Sciences, Hyderabad
| |
Collapse
|
228
|
Xiao G, Liu Y, Chen Y, He Z, Wen Y, Hu M. The Development and Validation of a Predictive Model for Voriconazole-Related Liver Injury in Hospitalized Patients in China. J Clin Med 2023; 12:4254. [PMID: 37445289 DOI: 10.3390/jcm12134254] [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: 04/19/2023] [Revised: 06/13/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Voriconazole is widely used in the treatment and prevention of invasive fungal diseases. Common drug-induced liver injuries increase the economic burdens and the risks of premature drug withdrawal and disease recurrence. This study estimated the disposal cost of voriconazole-related liver injury, explored the risk factors of voriconazole-related liver injury in hospitalized patients, and established a predictive model of liver injury to assist clinicians and pharmacists in estimating the probability or risk of liver injury after voriconazole administration to allow for early identification and intervention in patients at high risk of liver injury. A retrospective study was conducted on the selected inpatients whose blood concentration of voriconazole was measured in the West China Hospital of Sichuan University from September 2016 to June 2020. The incidence and disposal cost of voriconazole-related liver injuries were calculated. The incidence of voriconazole-related liver injury was 15.82% (217/1372). The disposal cost has been converted to 2023 at a discount rate of 5%. The median (P25, P75) disposal cost of severe liver injury (n = 42), general liver injury (n = 175), and non-liver injury (n = 1155) was 993.59 (361.70, 1451.76) Chinese yuan, 0.00 (0.00, 410.48) yuan, and 0.00 (0.00, 0.00) yuan, respectively, with a statistically significant difference (p < 0.001). Single factor analysis and multiple factor logistic regression were used to analyze the risk factors of voriconazole-related liver injury. The voriconazole-related liver injury was related to the trough concentration (Cmin, OR 1.099, 95% CI 1.058-1.140), hypoproteinemia (OR 1.723, 95% CI 1.126-2.636), and transplantation status (OR 0.555, 95% CI 0.325-0.948). The prediction model of liver injury was Logit (P)= -2.219 + 0.094 × Cmin + 0.544 × Hydroproteinemia - 0.589 × Transplantation, and the prediction model nomogram was established. The model validation results showed that the C-index of the derivation set and validation set was 0.706 and 0.733, respectively. The area under the curve (AUC) of the receiver operating characteristic (ROC) curve was 0.705 and 0.733, respectively, indicating that the model had good prediction ability. The prediction model will be helpful to develop clinical individualized medication of voriconazole and to identify and intervene in the cases of patients at high risk of voriconazole-related liver injury early on, in order to reduce the incidence of voriconazole-related liver injuries and the cost of treatment.
Collapse
Affiliation(s)
- Guirong Xiao
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yiyao Liu
- West China School of Pharmacy, Sichuan University, Chengdu 610041, China
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yanhua Chen
- Department of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Zhiyao He
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yan Wen
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ming Hu
- West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
229
|
Tonutti A, Simonetta E, Stainer A, Suigo G, De Santis M, Selmi C, Masetti C, Lleo A, Terracciano LM, Aliberti S, Amati F. Hepatic and pulmonary involvement in a patient with PR3-ANCA vasculitis following SARS-CoV-2 vaccination: A case report. Mod Rheumatol Case Rep 2023; 7:440-443. [PMID: 36695552 DOI: 10.1093/mrcr/rxad005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/21/2022] [Accepted: 01/11/2023] [Indexed: 01/26/2023]
Abstract
We here report the first case of anti-proteinase 3-positive anti-neutrophil cytoplasmic antibodies (ANCA)-associated vasculitis following the severe acute respiratory syndrome coronavirus 2 Pfizer-BioNTech vaccine presenting with prominent liver involvement and alveolar haemorrhage. Two weeks after vaccination, a 49-year-old man developed inflammatory arthralgias and hypertransaminasaemia. Two months later, fever and haemoptysis appeared; the patient tested positive for anti-proteinase 3 autoantibodies. High-dose steroids and rituximab were started, and complete remission was achieved. Systemic autoimmune diseases, including ANCA-associated vasculitis, should always be considered in the differential diagnosis of hypertransaminasaemia, especially when the clinical context is suspicious.
Collapse
Affiliation(s)
- Antonio Tonutti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Italy
| | | | - Anna Stainer
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Respiratory Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Giulia Suigo
- Respiratory Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Maria De Santis
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Carlo Selmi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Chiara Masetti
- Liver Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Ana Lleo
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Liver Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Luigi Maria Terracciano
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Institute of Pathology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Stefano Aliberti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Respiratory Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Francesco Amati
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Respiratory Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| |
Collapse
|
230
|
Weltzsch JP, Ziegler A, Lohse A. [Autoimmune hepatitis : From autoantibodies to cirrhosis]. INNERE MEDIZIN (HEIDELBERG, GERMANY) 2023:10.1007/s00108-023-01519-9. [PMID: 37306752 DOI: 10.1007/s00108-023-01519-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Accepted: 04/12/2023] [Indexed: 06/13/2023]
Abstract
Autoimmune Hepatitis (AIH) is an immune-mediated liver disease of unknown origin. Its clinical presentation is heterogeneous and ranges from asymptomatic courses over several years to acute forms with acute liver failure. Accordingly, the diagnosis is only made at the stage of cirrhosis in about one third of affected individuals. Early diagnosis and a consistent, adequate, individualized, immunosuppressive therapy are crucial for the prognosis, which is excellent when treated properly. AIH is rare in the general population and can be easily overlooked due to its variable clinical picture and sometimes difficult diagnosis. AIH should be considered as a differential diagnosis in any unclear acute or chronic hepatopathy. The therapy initially consists of remission induction and subsequently maintenance therapy with (often lifelong) immunosuppressants.
Collapse
Affiliation(s)
- Jan Philipp Weltzsch
- I. Med. Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Deutschland.
| | - Annerose Ziegler
- I. Med. Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Deutschland
| | - Ansgar Lohse
- I. Med. Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Deutschland
| |
Collapse
|
231
|
Hwang DB, Seo Y, Lee E, Won DH, Kim C, Kang M, Jeon Y, Kim HS, Park JW, Yun JW. Diagnostic potential of serum miR-532-3p as a circulating biomarker for experimental intrinsic drug-induced liver injury by acetaminophen and cisplatin in rats. Food Chem Toxicol 2023:113890. [PMID: 37308052 DOI: 10.1016/j.fct.2023.113890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 06/14/2023]
Abstract
Evaluating tissue injury largely depends on serum biochemical analysis despite insufficient tissue specificity and low sensitivity. Therefore, attention has been paid to the potential of microRNAs (miRNAs) to overcome the limitations of the current diagnostic tools, as tissue-enriched miRNAs are detected in the blood upon tissue injury. First, using a cisplatin-injected rats, we screened a specific pattern of altered hepatic miRNAs and their target mRNAs. Subsequently, we identified novel liver-specific circulating miRNAs for drug-induced liver injury by comparing miRNA expression changes in organs and serum. RNA sequencing revealed that 32 hepatic miRNAs were differentially expressed (DE) in the cisplatin-treated group. Furthermore, among the 1217 targets predicted using miRDB on these DE-miRNAs, 153 hepatic genes involved in different liver function-related pathways and processes were found to be dysregulated by cisplatin. Next, comparative analyses of the liver, kidneys, and serum DE-miRNAs were conducted to select circulating miRNA biomarker candidates reflecting drug-induced liver injury. Finally, among the four liver-specific circulating miRNAs selected based on their expression patterns in tissue and serum, miR-532-3p was increased in the serum after cisplatin or acetaminophen administration. Our findings suggest that miR-532-3p is potential as a serum biomarker for identifying drug-induced liver injury, leading to the accurate diagnosis.
Collapse
Affiliation(s)
- Da-Bin Hwang
- Department of Biotechnology, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Yoojin Seo
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Eunji Lee
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dong-Hoon Won
- Department of Biotechnology, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Changuk Kim
- Department of Biotechnology, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - MinHwa Kang
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Young Jeon
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyung-Sik Kim
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Jun Won Park
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Jun-Won Yun
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
232
|
Rao M, Nassiri V, Alhambra C, Snoeys J, Van Goethem F, Irrechukwu O, Aleo MD, Geys H, Mitra K, Will Y. AI/ML Models to Predict the Severity of Drug-Induced Liver Injury for Small Molecules. Chem Res Toxicol 2023. [PMID: 37294641 DOI: 10.1021/acs.chemrestox.3c00098] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Drug-induced liver injury (DILI), believed to be a multifactorial toxicity, has been a leading cause of attrition of small molecules during discovery, clinical development, and postmarketing. Identification of DILI risk early reduces the costs and cycle times associated with drug development. In recent years, several groups have reported predictive models that use physicochemical properties or in vitro and in vivo assay endpoints; however, these approaches have not accounted for liver-expressed proteins and drug molecules. To address this gap, we have developed an integrated artificial intelligence/machine learning (AI/ML) model to predict DILI severity for small molecules using a combination of physicochemical properties and off-target interactions predicted in silico. We compiled a data set of 603 diverse compounds from public databases. Among them, 164 were categorized as Most DILI (M-DILI), 245 as Less DILI (L-DILI), and 194 as No DILI (N-DILI) by the FDA. Six machine learning methods were used to create a consensus model for predicting the DILI potential. These methods include k-nearest neighbor (k-NN), support vector machine (SVM), random forest (RF), Naïve Bayes (NB), artificial neural network (ANN), logistic regression (LR), weighted average ensemble learning (WA) and penalized logistic regression (PLR). Among the analyzed ML methods, SVM, RF, LR, WA, and PLR identified M-DILI and N-DILI compounds, achieving a receiver operating characteristic area under the curve of 0.88, sensitivity of 0.73, and specificity of 0.9. Approximately 43 off-targets, along with physicochemical properties (fsp3, log S, basicity, reactive functional groups, and predicted metabolites), were identified as significant factors in distinguishing between M-DILI and N-DILI compounds. The key off-targets that we identified include: PTGS1, PTGS2, SLC22A12, PPARγ, RXRA, CYP2C9, AKR1C3, MGLL, RET, AR, and ABCC4. The present AI/ML computational approach therefore demonstrates that the integration of physicochemical properties and predicted on- and off-target biological interactions can significantly improve DILI predictivity compared to chemical properties alone.
Collapse
Affiliation(s)
- Mohan Rao
- Discovery, Product Development and Supply (DPDS), Preclinical Sciences and Translational Safety (PSTS), Predictive Investigative and Translational Toxicology (PITT), Janssen Pharmaceutical Companies of Johnson and Johnson, La Jolla, California 92121, United States
| | - Vahid Nassiri
- Open Analytics, Jupiterstraat 20, 2600 Antwerpen, Belgium
| | - Cristóbal Alhambra
- Discovery, Product Development and Supply (DPDS), Preclinical Sciences and Translational Safety (PSTS), Predictive Investigative and Translational Toxicology (PITT), Janssen Pharmaceutical Companies of Johnson and Johnson, La Jolla, California 92121, United States
| | - Jan Snoeys
- Discovery, Product Development and Supply (DPDS), Preclinical Sciences and Translational Safety (PSTS), Predictive Investigative and Translational Toxicology (PITT), Janssen Pharmaceutical Companies of Johnson and Johnson, La Jolla, California 92121, United States
| | - Freddy Van Goethem
- Discovery, Product Development and Supply (DPDS), Preclinical Sciences and Translational Safety (PSTS), Predictive Investigative and Translational Toxicology (PITT), Janssen Pharmaceutical Companies of Johnson and Johnson, La Jolla, California 92121, United States
| | - Onyi Irrechukwu
- Discovery, Product Development and Supply (DPDS), Preclinical Sciences and Translational Safety (PSTS), Predictive Investigative and Translational Toxicology (PITT), Janssen Pharmaceutical Companies of Johnson and Johnson, La Jolla, California 92121, United States
| | - Michael D Aleo
- TOXinsights LLC, Boiling Springs, Pennsylvania 17007, United States
| | - Helena Geys
- Discovery, Product Development and Supply (DPDS), Preclinical Sciences and Translational Safety (PSTS), Predictive Investigative and Translational Toxicology (PITT), Janssen Pharmaceutical Companies of Johnson and Johnson, La Jolla, California 92121, United States
| | - Kaushik Mitra
- Discovery, Product Development and Supply (DPDS), Preclinical Sciences and Translational Safety (PSTS), Predictive Investigative and Translational Toxicology (PITT), Janssen Pharmaceutical Companies of Johnson and Johnson, La Jolla, California 92121, United States
| | - Yvonne Will
- Discovery, Product Development and Supply (DPDS), Preclinical Sciences and Translational Safety (PSTS), Predictive Investigative and Translational Toxicology (PITT), Janssen Pharmaceutical Companies of Johnson and Johnson, La Jolla, California 92121, United States
| |
Collapse
|
233
|
Yamamoto T, Mizuno K, Ito T, Yokoyama S, Yamamoto K, Imai N, Ishizu Y, Honda T, Ishikawa T, Kanamori A, Yasuda S, Toyoda H, Yokota K, Hase T, Nishio N, Maeda O, Ishii M, Sone M, Ando Y, Akiyama M, Ishigami M, Kawashima H. Abdominal pain accompanied by elevated serum inflammatory markers and biliary enzymes for diagnosing immune checkpoint inhibitor-induced sclerosing cholangitis. Invest New Drugs 2023; 41:512-521. [PMID: 37171720 DOI: 10.1007/s10637-023-01366-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 04/24/2023] [Indexed: 05/13/2023]
Abstract
Immune-related sclerosing cholangitis (irSC) is relatively rare and its clinical characteristics are not well known. In this study, we aimed to summarize the clinical features of irSC. Clinical data were collected retrospectively from 1,393 patients with advanced malignancy treated with immune-checkpoint inhibitors (ICIs) between August 2014 and October 2021. We analyzed patients with immune-related adverse events of liver injury (liver-irAEs) and compared irSC and non-irSC groups. Sixty-seven patients (4.8%) had a liver-irAE (≥ grade 3) during the follow-up period (median, 262 days). Among these, irSC was observed in eight patients (11.9%). All patients in the irSC group were treated with anti-PD-1/PD-L1 antibodies. Compared with the non-irSC group, the irSC group showed mainly non-hepatocellular liver injury (87.5 % vs 50.8 %, P = 0.065), and had elevated serum inflammatory markers (e.g., CRP and NLR) and biliary enzymes (e.g., GGTP and ALP) at the onset of liver-irAEs. Furthermore, most patients with irSC had abdominal pain. In the non-irSC group, the liver injury of 23 patients improved only with the discontinuation of ICIs, and 22 patients improved with medication including prednisolone (PSL). Conversely, almost all patients (n=7) in the irSC group were treated with PSL, but only two patients experienced an improvement in liver injury. We found that irSC is characterized by a non-hepatocellular type of liver injury with abdominal pain and a high inflammatory response and is refractory to treatment. Further examination by imaging is recommended to detect intractable irSC in cases with these characteristics.
Collapse
Affiliation(s)
- Takafumi Yamamoto
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuyuki Mizuno
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takanori Ito
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Shinya Yokoyama
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kenta Yamamoto
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Norihiro Imai
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoji Ishizu
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Honda
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takuya Ishikawa
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akira Kanamori
- Department of Gastroenterology and Hepatology, Ogaki Municipal Hospital, Ogaki, Japan
| | - Satoshi Yasuda
- Department of Gastroenterology and Hepatology, Ogaki Municipal Hospital, Ogaki, Japan
| | - Hidenori Toyoda
- Department of Gastroenterology and Hepatology, Ogaki Municipal Hospital, Ogaki, Japan
| | - Kenji Yokota
- Department of Dermatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tetsunari Hase
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Naoki Nishio
- Department of Otorhinolaryngology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Osamu Maeda
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Japan
| | - Makoto Ishii
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Michihiko Sone
- Department of Otorhinolaryngology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuichi Ando
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Japan
| | - Masashi Akiyama
- Department of Dermatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masatoshi Ishigami
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroki Kawashima
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
234
|
Hountondji L, Ferreira De Matos C, Lebossé F, Quantin X, Lesage C, Palassin P, Rivet V, Faure S, Pageaux GP, Assenat É, Alric L, Zahhaf A, Larrey D, Witkowski Durand Viel P, Riviere B, Janick S, Dalle S, Maria ATJ, Comont T, Meunier L. Clinical pattern of checkpoint inhibitor-induced liver injury in a multicentre cohort. JHEP Rep 2023; 5:100719. [PMID: 37138674 PMCID: PMC10149360 DOI: 10.1016/j.jhepr.2023.100719] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/10/2023] [Accepted: 02/16/2023] [Indexed: 05/05/2023] Open
Abstract
Background & Aims Immune checkpoint inhibitors (ICIs) have changed the landscape of cancer therapy. Liver toxicity occurs in up to 25% of patients treated with ICIs. The aim of our study was to describe the different clinical patterns of ICI-induced hepatitis and to assess their outcome. Methods We conducted a retrospective observational study of patients with checkpoint inhibitor-induced liver injury (CHILI) discussed in multidisciplinary meetings between December 2018 and March 2022 in three French centres specialised in ICI toxicity management (Montpellier, Toulouse, Lyon). The hepatitis clinical pattern was analysed by the ratio of serum alanine aminotransferase (ALT) and alkaline phosphatase (ALP) (R value = (ALT/ULN)/(ALP/ULN)) for characterisation as cholestatic (R ≤2), hepatocellular (R ≥5), or mixed (2 <R <5). Results We included 117 patients with CHILI. The clinical pattern was hepatocellular in 38.5%, cholestatic in 36.8%, and mixed in 24.8% of patients. High-grade hepatitis severity (grade ≥3 according to the Common Terminology Criteria for Adverse Events system) was significantly associated with the hepatocellular hepatitis (p <0.05). No cases of severe acute hepatitis were reported. Liver biopsy was performed in 41.9% of patients: granulomatous lesions, endothelitis, or lymphocytic cholangitis were described. Biliary stenosis occurred in eight patients (6.8%) and was significantly more frequent in the cholestatic clinical pattern (p < 0.001). Steroids alone were mainly administered to patients with a hepatocellular clinical pattern (26.5%), and ursodeoxycholic acid was more frequently used in the cholestatic pattern (19.7%) than in the hepatocellular or mixed clinical pattern (p <0.001). Seventeen patients improved without any treatment. Among the 51 patients (43.6%) rechallenged with ICIs, 12 (23.5%) developed CHILI recurrence. Conclusions This large cohort indicates the different clinical patterns of ICI-induced liver injury and highlights that the cholestatic and hepatocellular patterns are the most frequent with different outcomes. Impact and Implications ICIs can induce hepatitis. In this retrospective series, we report 117 cases of ICI-induced hepatitis, mostly grades 3 and 4. We find a similar distribution of the different patterns of hepatitis. ICI could be resumed without systematic recurrence of hepatitis.
Collapse
Affiliation(s)
- Lina Hountondji
- Department of Liver Transplantation, Saint Eloi Hospital, Montpellier University Hospital, Montpellier, France
| | | | - Fanny Lebossé
- Department of Hepatology, Croix Rousse Hospital, Lyon Liver Institute, Hospices Civils of Lyon, Lyon, France
| | - Xavier Quantin
- Department of Medical Oncology, Montpellier Cancer Institute, Montpellier University Hospital, Montpellier, France
| | - Candice Lesage
- Department of Dermatology, Saint Eloi Hospital, Montpellier University Hospital, Montpellier, France
| | - Pascale Palassin
- Department of Medical Pharmacology and Toxicology, Lapeyronie Hospital, Montpellier University Hospital, Montpellier, France
| | - Valérian Rivet
- Department of Internal Medicine, IUCT-Oncopole, Toulouse University Hospital, Toulouse, France
| | - Stéphanie Faure
- Department of Liver Transplantation, Saint Eloi Hospital, Montpellier University Hospital, Montpellier, France
| | - Georges-Philippe Pageaux
- Department of Liver Transplantation, Saint Eloi Hospital, Montpellier University Hospital, Montpellier, France
| | - Éric Assenat
- Department of Oncology, Saint Eloi Hospital, Montpellier University Hospital, Montpellier, France
| | - Laurent Alric
- Department of Internal Medicine and Digestive Diseases, Purpan Hospital, Toulouse University Hospital, Toulouse, France
| | - Amel Zahhaf
- Department of Liver Transplantation, Saint Eloi Hospital, Montpellier University Hospital, REFHEPS, Montpellier, France
| | - Dominique Larrey
- Department of Liver Transplantation, Saint Eloi Hospital, Montpellier University Hospital, REFHEPS, Montpellier, France
| | | | - Benjamin Riviere
- Department of Pathology, Montpellier University Hospital, University of Montpellier, Montpellier, France
| | | | - Stéphane Dalle
- Department of Dermatology, Lyon Sud Hospital, Lyon Cancer Institute, Hospices Civils of Lyon, Lyon, France
| | - Alexandre Thibault Jacques Maria
- Internal Medicine & Immuno-Oncology (MedI2O), Institute for Regenerative Medicine and Biotherapy (IRMB), Saint Eloi Hospital, Montpellier University Hospital, Montpellier, France
| | - Thibaut Comont
- Department of Internal Medicine, IUCT-Oncopole, Toulouse University Hospital, Toulouse, France
| | - Lucy Meunier
- Department of Liver Transplantation, Saint Eloi Hospital, Montpellier University Hospital, REFHEPS, Montpellier, France
- Corresponding author. Address: Department of Liver Transplantation, Saint Eloi Hospital, Montpellier University Hospital, REFHEPS, 80 avenue Augustin Fliche, 34090 Montpellier, France. Tel: +33 4 67 33 02 24, Fax: +33 4 67 33 69 42.
| |
Collapse
|
235
|
Zeng G, Eslick GD, Weltman M. Systematic review and meta-analysis: Comparing hepatocellular and cholestatic patterns of drug-induced liver injury. ILIVER 2023; 2:122-129. [DOI: 10.1016/j.iliver.2023.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
|
236
|
Chen C, Gong W, Tian J, Gao X, Qin X, Du G, Zhou Y. Radix Paeoniae Alba attenuates Radix Bupleuri-induced hepatotoxicity by modulating gut microbiota to alleviate the inhibition of saikosaponins on glutathione synthetase. J Pharm Anal 2023; 13:640-659. [PMID: 37440914 PMCID: PMC10334278 DOI: 10.1016/j.jpha.2023.04.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/07/2023] [Accepted: 04/23/2023] [Indexed: 07/15/2023] Open
Abstract
Radix Bupleuri (RB) is commonly used to treat depression, but it can also lead to hepatotoxicity after long-term use. In many anti-depression prescriptions, RB is often used in combination with Radix Paeoniae Alba (RPA) as an herb pair. However, whether RPA can alleviate RB-induced hepatotoxicity remain unclear. In this work, the results confirmed that RB had a dose-dependent antidepressant effect, but the optimal antidepressant dose caused hepatotoxicity. Notably, RPA effectively reversed RB-induced hepatotoxicity. Afterward, the mechanism of RB-induced hepatotoxicity was confirmed. The results showed that saikosaponin A and saikosaponin D could inhibit GSH synthase (GSS) activity in the liver, and further cause liver injury through oxidative stress and nuclear factor kappa B (NF-κB)/NOD-like receptor thermal protein domain associated protein 3 (NLRP3) pathway. Furthermore, the mechanisms by which RPA attenuates RB-induced hepatotoxicity were investigated. The results demonstrated that RPA increased the abundance of intestinal bacteria with glycosidase activity, thereby promoting the conversion of saikosaponins to saikogenins in vivo. Different from saikosaponin A and saikosaponin D, which are directly combined with GSS as an inhibitor, their deglycosylation conversion products saikogenin F and saikogenin G exhibited no GSS binding activity. Based on this, RPA can alleviate the inhibitory effect of saikosaponins on GSS activity to reshape the liver redox balance and further reverse the RB-induced liver inflammatory response by the NF-κB/NLRP3 pathway. In conclusion, the present study suggests that promoting the conversion of saikosaponins by modulating gut microbiota to attenuate the inhibition of GSS is the potential mechanism by which RPA prevents RB-induced hepatotoxicity.
Collapse
Affiliation(s)
- Congcong Chen
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, 030006, China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, Taiyuan, 030006, China
| | - Wenxia Gong
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, 030006, China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, Taiyuan, 030006, China
| | - Junshen Tian
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, 030006, China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, Taiyuan, 030006, China
| | - Xiaoxia Gao
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, 030006, China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, Taiyuan, 030006, China
| | - Xuemei Qin
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, 030006, China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, Taiyuan, 030006, China
| | - Guanhua Du
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, 030006, China
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yuzhi Zhou
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, 030006, China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, Taiyuan, 030006, China
| |
Collapse
|
237
|
Stern S, Wang H, Sadrieh N. Microphysiological Models for Mechanistic-Based Prediction of Idiosyncratic DILI. Cells 2023; 12:1476. [PMID: 37296597 PMCID: PMC10253021 DOI: 10.3390/cells12111476] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/18/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Drug-induced liver injury (DILI) is a major contributor to high attrition rates among candidate and market drugs and a key regulatory, industry, and global health concern. While acute and dose-dependent DILI, namely, intrinsic DILI, is predictable and often reproducible in preclinical models, the nature of idiosyncratic DILI (iDILI) limits its mechanistic understanding due to the complex disease pathogenesis, and recapitulation using in vitro and in vivo models is extremely challenging. However, hepatic inflammation is a key feature of iDILI primarily orchestrated by the innate and adaptive immune system. This review summarizes the in vitro co-culture models that exploit the role of the immune system to investigate iDILI. Particularly, this review focuses on advancements in human-based 3D multicellular models attempting to supplement in vivo models that often lack predictability and display interspecies variations. Exploiting the immune-mediated mechanisms of iDILI, the inclusion of non-parenchymal cells in these hepatoxicity models, namely, Kupffer cells, stellate cells, dendritic cells, and liver sinusoidal endothelial cells, introduces heterotypic cell-cell interactions and mimics the hepatic microenvironment. Additionally, drugs recalled from the market in the US between 1996-2010 that were studies in these various models highlight the necessity for further harmonization and comparison of model characteristics. Challenges regarding disease-related endpoints, mimicking 3D architecture with different cell-cell contact, cell source, and the underlying multi-cellular and multi-stage mechanisms are described. It is our belief that progressing our understanding of the underlying pathogenesis of iDILI will provide mechanistic clues and a method for drug safety screening to better predict liver injury in clinical trials and post-marketing.
Collapse
Affiliation(s)
- Sydney Stern
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD 21201, USA;
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD 21201, USA;
| | - Nakissa Sadrieh
- Office of New Drugs, Center of Drug Evaluation and Research, FDA, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| |
Collapse
|
238
|
Song Q, Mao X, Jing M, Fu Y, Yan W. Pathophysiological role of BACH transcription factors in digestive system diseases. Front Physiol 2023; 14:1121353. [PMID: 37228820 PMCID: PMC10203417 DOI: 10.3389/fphys.2023.1121353] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 04/26/2023] [Indexed: 05/27/2023] Open
Abstract
BTB and CNC homologous (BACH) proteins, including BACH1 and BACH2, are transcription factors that are widely expressed in human tissues. BACH proteins form heterodimers with small musculoaponeurotic fibrosarcoma (MAF) proteins to suppress the transcription of target genes. Furthermore, BACH1 promotes the transcription of target genes. BACH proteins regulate physiological processes, such as the differentiation of B cells and T cells, mitochondrial function, and heme homeostasis as well as pathogenesis related to inflammation, oxidative-stress damage caused by drugs, toxicants, or infections; autoimmunity disorders; and cancer angiogenesis, epithelial-mesenchymal transition, chemotherapy resistance, progression, and metabolism. In this review, we discuss the function of BACH proteins in the digestive system, including the liver, gallbladder, esophagus, stomach, small and large intestines, and pancreas. BACH proteins directly target genes or indirectly regulate downstream molecules to promote or inhibit biological phenomena such as inflammation, tumor angiogenesis, and epithelial-mesenchymal transition. BACH proteins are also regulated by proteins, miRNAs, LncRNAs, labile iron, and positive and negative feedback. Additionally, we summarize a list of regulators targeting these proteins. Our review provides a reference for future studies on targeted drugs in digestive diseases.
Collapse
Affiliation(s)
- Qianben Song
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xin Mao
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Mengjia Jing
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yu Fu
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Yan
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
239
|
Carrascosa JM, Vilarrasa E, Belinchón I, Herranz P, Crespo J, Guimerá F, Olveira A. [Translated article] Common Approach to Metabolic-Associated Fatty Liver Disease in Patients With Psoriasis: Consensus-Based Recommendations From a Multidisciplinary Group of Experts. ACTAS DERMO-SIFILIOGRAFICAS 2023; 114:T392-T401. [PMID: 37068635 DOI: 10.1016/j.ad.2023.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/17/2023] [Indexed: 04/19/2023] Open
Abstract
Recent years have seen concerted efforts to understand the relation between psoriasis and metabolic-associated fatty liver disease (MAFLD). Not only is MALFD diagnosed more often in patients with psoriasis, but its clinical course is also more aggressive. A common approach is therefore needed to enable early detection of liver disease coincident with psoriasis. Especially important is an analysis of risks and benefits of potentially hepatotoxic treatments. This consensus paper presents the recommendations of a group of experts in dermatology and hepatology regarding screening for MALFD as well as criteria for monitoring patients and referring them to hepatologists when liver disease is suspected.
Collapse
Affiliation(s)
- J M Carrascosa
- Departamento de Dermatología, Hospital Universitario Germans Trias i Pujol, Universitat Autònoma de Barcelona, IGTP Badalona, Barcelona, Spain.
| | - E Vilarrasa
- Departamento de Dermatología, Hospital Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - I Belinchón
- Departamento de Dermatología, Hospital General Universitario de Alicante, Instituto de Investigación Sanitaria y Biomédica (ISABIAL), Universidad Miguel Hernández de Elche, Alicante, Spain
| | - P Herranz
- Departamento de Dermatología, Hospital Universitario La Paz, Madrid, Spain
| | - J Crespo
- Servicio de Gastroenterología y Hepatología, Hospital Universitario Marqués de Valdecilla, IDIVAL, Escuela de Medicina, Universidad de Cantabria, Santander, Spain
| | - F Guimerá
- Servicio de Dermatología y Patología, Hospital Universitario de Canarias, La Laguna, Spain
| | - A Olveira
- Servicio de Aparato Digestivo, Hospital Universitario La Paz, Madrid, Spain
| |
Collapse
|
240
|
Zhang CJ, Meyer SR, O’Meara MJ, Huang S, Capeling MM, Ferrer-Torres D, Childs CJ, Spence JR, Fontana RJ, Sexton JZ. A human liver organoid screening platform for DILI risk prediction. J Hepatol 2023; 78:998-1006. [PMID: 36738840 PMCID: PMC11268729 DOI: 10.1016/j.jhep.2023.01.019] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Drug-induced liver injury (DILI), both intrinsic and idiosyncratic, causes frequent morbidity, mortality, clinical trial failures and post-approval withdrawal. This suggests an unmet need for improved in vitro models for DILI risk prediction that can account for diverse host genetics and other clinical factors. In this study, we evaluated the utility of human liver organoids (HLOs) for high-throughput DILI risk prediction and in an organ-on-chip system. METHODS HLOs were derived from three separate iPSC lines and benchmarked on two platforms for their ability to model in vitro liver function and identify hepatotoxic compounds using biochemical assays for albumin, ALT, AST, microscopy-based morphological profiling, and single-cell transcriptomics: i) HLOs dispersed in 384-well-formatted plates and exposed to a library of compounds; ii) HLOs adapted to a liver-on-chip system. RESULTS Dispersed HLOs derived from the three iPSC lines had similar DILI predictive capacity as intact HLOs in a high-throughput screening format, allowing for measurable IC50 values of compound cytotoxicity. Distinct morphological differences were observed in cells treated with drugs exerting differing mechanisms of toxicity. On-chip HLOs significantly increased albumin production, CYP450 expression, and ALT/AST release when treated with known hepatoxic drugs compared to dispersed HLOs and primary human hepatocytes. On-chip HLOs were able to predict the synergistic hepatotoxicity of tenofovir-inarigivir and displayed steatosis and mitochondrial perturbation, via phenotypic and transcriptomic analysis, on exposure to fialuridine and acetaminophen, respectively. CONCLUSIONS The high-throughput and liver-on-chip systems exhibit enhanced in vivo-like functions and demonstrate the potential utility of these platforms for DILI risk assessment. Tenofovir-inarigivr-associated hepatotoxicity was observed and correlates with the clinical manifestation of DILI observed in patients. IMPACT AND IMPLICATIONS Idiosyncratic (spontaneous, patient-specific) drug-induced liver injury (DILI) is difficult to study due to the lack of liver models that function as human liver tissue and are adaptable for large-scale drug screening. Human liver organoids grown from patient stem cells respond to known DILI-causing drugs in both a high-throughput and on a physiological "chip" culture system. These platforms show promise for researchers in their use as predictive models for novel drugs before entering clinical trials and as a potential in vitro diagnostic tool. Our findings support further development of patient-derived liver organoid lines and their use in the context of DILI research.
Collapse
Affiliation(s)
- Charles J. Zhang
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Sophia R. Meyer
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Matthew J. O’Meara
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Sha Huang
- Department of Internal Medicine, Gastroenterology and Hepatology, Michigan Medicine at the University of Michigan, Ann Arbor, MI, 48109, USA
| | - Meghan M. Capeling
- Department of Internal Medicine, Gastroenterology and Hepatology, Michigan Medicine at the University of Michigan, Ann Arbor, MI, 48109, USA
| | - Daysha Ferrer-Torres
- Department of Internal Medicine, Gastroenterology and Hepatology, Michigan Medicine at the University of Michigan, Ann Arbor, MI, 48109, USA
| | - Charlie J. Childs
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jason R. Spence
- Department of Internal Medicine, Gastroenterology and Hepatology, Michigan Medicine at the University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Robert J. Fontana
- Department of Internal Medicine, Gastroenterology and Hepatology, Michigan Medicine at the University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jonathan Z. Sexton
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, Gastroenterology and Hepatology, Michigan Medicine at the University of Michigan, Ann Arbor, MI, 48109, USA
- U-M Center for Drug Repurposing, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
241
|
Carrascosa JM, Vilarrasa E, Belinchón I, Herranz P, Crespo J, Guimerá F, Olveira A. Common Approach to Metabolic-Associated Fatty Liver Disease in Patients With Psoriasis: Consensus-Based Recommendations From a Multidisciplinary Group of Experts. ACTAS DERMO-SIFILIOGRAFICAS 2023; 114:392-401. [PMID: 36720362 DOI: 10.1016/j.ad.2023.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/11/2023] [Accepted: 01/17/2023] [Indexed: 01/31/2023] Open
Abstract
Recent years have seen concerted efforts to understand the relation between psoriasis and metabolic-associated fatty liver disease (MAFLD). Not only is MALFD diagnosed more often in patients with psoriasis, but its clinical course is also more aggressive. A common approach is therefore needed to enable early detection of liver disease coincident with psoriasis. Especially important is an analysis of risks and benefits of potentially hepatotoxic treatments. This consensus paper presents the recommendations of a group of experts in dermatology and hepatology regarding screening for MALFD as well as criteria for monitoring patients and referring them to hepatologists when liver disease is suspected.
Collapse
Affiliation(s)
- J M Carrascosa
- Departamento de Dermatología, Hospital Universitario Germans Trias i Pujol, Universitat Autònoma de Barcelona. IGTP Badalona, Barcelona, España.
| | - E Vilarrasa
- Departamento de Dermatología, Hospital Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, España
| | - I Belinchón
- Departamento de Dermatología, Hospital General Universitario de Alicante, Instituto de Investigación Sanitaria y Biomédica (ISABIAL), Universidad Miguel Hernández de Elche, Alicante, España
| | - P Herranz
- Departamento de Dermatología, Hospital Universitario La Paz, Madrid, España
| | - J Crespo
- Servicio de Gastroenterología y Hepatología, Hospital Universitario Marqués de Valdecilla. IDIVAL. Escuela de Medicina. Universidad de Cantabria, Santander, España
| | - F Guimerá
- Servicio de Dermatología y Patología, Hospital Universitario de Canarias, La Laguna, España
| | - A Olveira
- Servicio de Aparato Digestivo, Hospital Universitario La Paz, Madrid, España
| |
Collapse
|
242
|
Liatsos GD. SARS-CoV-2 induced liver injury: Incidence, risk factors, impact on COVID-19 severity and prognosis in different population groups. World J Gastroenterol 2023; 29:2397-2432. [PMID: 37179584 PMCID: PMC10167898 DOI: 10.3748/wjg.v29.i16.2397] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/17/2023] [Accepted: 04/07/2023] [Indexed: 04/24/2023] Open
Abstract
Liver is unlikely the key organ driving mortality in coronavirus disease 2019 (COVID-19) however, liver function tests (LFTs) abnormalities are widely observed mostly in moderate and severe cases. According to this review, the overall prevalence of abnormal LFTs in COVID-19 patients ranges from 2.5% to 96.8% worldwide. The geographical variability in the prevalence of underlying diseases is the determinant for the observed discrepancies between East and West. Multifactorial mechanisms are implicated in COVID-19-induced liver injury. Among them, hypercytokinemia with "bystander hepatitis", cytokine storm syndrome with subsequent oxidative stress and endotheliopathy, hypercoagulable state and immuno-thromboinflammation are the most determinant mechanisms leading to tissue injury. Liver hypoxia may also contribute under specific conditions, while direct hepatocyte injury is an emerging mechanism. Except for initially observed severe acute respiratory distress syndrome corona virus-2 (SARS-CoV-2) tropism for cholangiocytes, more recent cumulative data show SARS-CoV-2 virions within hepatocytes and sinusoidal endothelial cells using electron microscopy (EM). The best evidence for hepatocellular invasion by the virus is the identification of replicating SARS-CoV-2 RNA, S protein RNA and viral nucleocapsid protein within hepatocytes using in-situ hybridization and immunostaining with observed intrahepatic presence of SARS-CoV-2 by EM and by in-situ hybridization. New data mostly derived from imaging findings indicate possible long-term sequelae for the liver months after recovery, suggesting a post-COVID-19 persistent live injury.
Collapse
Affiliation(s)
- George D Liatsos
- Department of Internal Medicine, Hippokration General Hospital, Athens 11527, Attiki, Greece
| |
Collapse
|
243
|
Huang D, Peng J, Lei L, Chen Y, Zhu Z, Cai Q, Deng Y, Chen J. Time of Liver Function Abnormal Identification on Prediction of the Risk of Anti-tuberculosis-induced Liver Injury. J Clin Transl Hepatol 2023; 11:425-432. [PMID: 36643044 PMCID: PMC9817055 DOI: 10.14218/jcth.2022.00077] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/16/2022] [Accepted: 04/25/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND AND AIMS Anti-tuberculosis (anti-TB) drug-induced liver injury (AT-DILI) is the most common side effect in patients who received anti-TB therapy. AT-DILI management includes monitoring liver function until symptoms arise in patients without high-risk factors for liver damage. The present study aimed to investigate the effect of liver function test (LFT) abnormal identification on the risk of DILI, including liver failure and anti-TB drug resistance in patients without high-risk factors. METHODS A total of 399 patients without high-risk factors for liver damage at baseline and who experienced LFT abnormal during the 6 months of first-line anti-TB treatment were enrolled. The Roussel Uclaf Causal Relationship Assessment Method (RUCAM, 2016) was applied in suspected DILI. The correlations between the time of LFT abnormal identification and DILI, liver failure, and anti-TB drug resistance were analyzed by smooth curve fitting and multivariable logistic regression models. RESULTS Among all study patients, 131 met the criteria for DILI with a mean RUCAM causality score of 8.86±0.63. 26/131 and 105/131 were in the probable grading and highly probable grading, respectively. The time of abnormal LFT identification was an independent predictor of DILI, liver failure, and anti-TB drug resistance in the crude model and after adjusting for other risk patient factors. The time of abnormal LFT identification was positively correlated with DILI, liver failure, and anti-TB drug resistance. The late identification group (>8 weeks) had the highest risk of DILI, followed by liver failure compared with the other two groups. CONCLUSIONS The time to identification of LFT was positively correlated with DILI, liver failure, and anti-TB drug resistance. The risk of DILI and liver failure was significantly increased in the late identification group with abnormal LFT identified after 8 weeks compared with 4 and 8 weeks. Early monitoring of LFT is recommended for patients without the high-risk factor of DILI after anti-TB treatment is initiated.
Collapse
Affiliation(s)
- Deliang Huang
- Department of Liver Diseases, The Third People’s Hospital of Shenzhen, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Jinghan Peng
- Department of Liver Diseases, The Third People’s Hospital of Shenzhen, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Lin Lei
- Department of Tuberculosis, The Third People’s Hospital of Shenzhen, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Yuanyuan Chen
- Department of Liver Diseases, The Third People’s Hospital of Shenzhen, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Zhibing Zhu
- Department of Liver Diseases, The Third People’s Hospital of Shenzhen, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Qingxian Cai
- Department of Liver Diseases, The Third People’s Hospital of Shenzhen, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Yongcong Deng
- Department of Tuberculosis, The Third People’s Hospital of Shenzhen, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Jun Chen
- Department of Liver Diseases, The Third People’s Hospital of Shenzhen, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
- Correspondence to: Jun Chen, Department of Liver Diseases, The Third People’s Hospital of Shenzhen, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong 518100, China. ORCID: https://orcid.org/0000-0002-9744-792X. Tel: +86-755-61222333, Fax: +86-755-61238928, E-mail:
| |
Collapse
|
244
|
Vincent ML, Kraft RM, Ratelle JT. 75-Year-Old Woman With Deranged Liver Enzymes. Mayo Clin Proc 2023; 98:784-788. [PMID: 37028982 DOI: 10.1016/j.mayocp.2022.09.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/20/2022] [Accepted: 09/22/2022] [Indexed: 04/09/2023]
Affiliation(s)
- Matthew L Vincent
- Resident in Internal Medicine, Mayo Clinic School of Graduate Medical Education, Rochester, MN
| | - Robert M Kraft
- Resident in Internal Medicine, Mayo Clinic School of Graduate Medical Education, Rochester, MN
| | - John T Ratelle
- Advisor to residents and Consultant in Hospital Internal Medicine, Mayo Clinic, Rochester, MN.
| |
Collapse
|
245
|
Izawa T, Travlos GS, Cortes RA, Clayton NP, Sills RC, Pandiri AR. Absence of Increased Susceptibility to Acetaminophen-Induced Liver Injury in a Diet-Induced NAFLD Mouse Model. Toxicol Pathol 2023; 51:112-125. [PMID: 37158481 PMCID: PMC10523943 DOI: 10.1177/01926233231171101] [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] [Indexed: 05/10/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a common chronic liver disease and its influence on drug-induced liver injury (DILI) is not fully understood. We investigated whether NAFLD can influence acetaminophen (APAP [N-acetyl-p-aminophenol])-induced hepatotoxicity in a diet-induced obese (DIO) mouse model of NAFLD. The male C57BL/6NTac DIO mice, fed a high-fat diet for more than 12 weeks, developed obesity, hyperinsulinemia, impaired glucose tolerance, and hepatomegaly with hepatic steatosis, similar to human NAFLD. In the acute toxicity study after a single dose of APAP (150 mg/kg), compared with control lean mice, the DIO mice had decreased serum transaminase levels and less severe hepatocellular injury. The DIO mice also had altered expression of genes related to APAP metabolism. Chronic APAP exposure for 26 weeks did not predispose the DIO mice with NAFLD to more severe hepatotoxicity compared with the lean mice. These results suggested that the C57BL/6NTac DIO mouse model appears to be more tolerant to APAP-induced hepatotoxicity than lean mice, potentially related to altered xenobiotic metabolizing capacity in the fatty liver. Further mechanistic studies with APAP and other drugs in NAFLD animal models are necessary to investigate the mechanism of altered susceptibility to intrinsic DILI in some human NAFLD patients.
Collapse
Affiliation(s)
- Takeshi Izawa
- Comparative and Molecular Pathogenesis Branch, Division of Translational Toxicology, NIEHS, Research Triangle Park, NC, USA
- Laboratory of Veterinary Pathology, Osaka Metropolitan University, Izumisano, Osaka, Japan
| | - Gregory S. Travlos
- Comparative and Molecular Pathogenesis Branch, Division of Translational Toxicology, NIEHS, Research Triangle Park, NC, USA
| | - Ricardo A. Cortes
- Comparative and Molecular Pathogenesis Branch, Division of Translational Toxicology, NIEHS, Research Triangle Park, NC, USA
- Experimental Pathology Laboratories, Inc., Research Triangle Park, NC, USA
| | - Natasha P. Clayton
- Comparative and Molecular Pathogenesis Branch, Division of Translational Toxicology, NIEHS, Research Triangle Park, NC, USA
| | - Robert C. Sills
- Comparative and Molecular Pathogenesis Branch, Division of Translational Toxicology, NIEHS, Research Triangle Park, NC, USA
| | - Arun R. Pandiri
- Comparative and Molecular Pathogenesis Branch, Division of Translational Toxicology, NIEHS, Research Triangle Park, NC, USA
| |
Collapse
|
246
|
Niu H, Ma J, Medina-Caliz I, Robles-Diaz M, Bonilla-Toyos E, Ghabril M, Lucena MI, Alvarez-Alvarez I, Andrade RJ. Potential benefit and lack of serious risk from corticosteroids in drug-induced liver injury: An international, multicentre, propensity score-matched analysis. Aliment Pharmacol Ther 2023; 57:886-896. [PMID: 36547393 DOI: 10.1111/apt.17373] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/29/2022] [Accepted: 12/11/2022] [Indexed: 03/24/2023]
Abstract
BACKGROUND The use of corticosteroids to treat patients with idiosyncratic drug-induced liver injury (DILI) relies on empirical clinical decisions. AIM To investigate the relationship between corticosteroids and risk of acute liver failure (ALF) in patients with DILI and to assess if corticosteroid therapy was associated with improved outcomes in DILI patients. METHODS We analysed bona fide idiosyncratic DILI cases from the Spanish DILI Registry and Indiana University School of Medicine. Patients treated with corticosteroids were compared to those who did not receive any treatment. Nearest neighbour propensity score matching analyses were conducted. RESULTS We enrolled 724 patients, 106 under corticosteroid therapy, in whom there was over-representation of more severe injury and autoimmune features, and 618 who did not receive any treatment. In an analysis of 80 pairs of propensity score-matched patients, corticosteroid administration was not associated with an increased risk of developing ALF (odds ratio = 0.65; 95% confidence interval [CI]: 0.18-2.40; p = 0.518). Furthermore, in an additional analysis, a Cox regression model that included 41 propensity score-matched pairs showed that patients receiving corticosteroids had a significantly higher normalisation rate of liver enzymes than untreated patients (hazard ratio [HR] = 1.84; 95% CI: 1.02-3.32; p = 0.043), particularly in patients with serious injury who did not resolve within 30 days (HR = 2.79; 95% CI: 1.20-6.50; p = 0.018). CONCLUSION Corticosteroid therapy did not worsen outcome in DILI patients. Indeed, corticosteroid administration was associated with a greater rate of normalisation of liver enzymes in patients with serious DILI.
Collapse
Affiliation(s)
- Hao Niu
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain
- Biomedical Research Network Center for Hepatic and Digestive Diseases (CIBERehd), Carlos III Health Institute, Madrid, Spain
- Platform for Clinical Research and Clinical Trials IBIMA, Plataforma ISCIII de Investigación Clínica, Madrid, Spain
| | - Jiayi Ma
- Division of Gastroenterology/Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Inmaculada Medina-Caliz
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain
| | - Mercedes Robles-Diaz
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain
- Biomedical Research Network Center for Hepatic and Digestive Diseases (CIBERehd), Carlos III Health Institute, Madrid, Spain
| | - Elvira Bonilla-Toyos
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain
- Platform for Clinical Research and Clinical Trials IBIMA, Plataforma ISCIII de Investigación Clínica, Madrid, Spain
| | - Marwan Ghabril
- Division of Gastroenterology/Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Mª Isabel Lucena
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain
- Biomedical Research Network Center for Hepatic and Digestive Diseases (CIBERehd), Carlos III Health Institute, Madrid, Spain
- Platform for Clinical Research and Clinical Trials IBIMA, Plataforma ISCIII de Investigación Clínica, Madrid, Spain
| | - Ismael Alvarez-Alvarez
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain
- Biomedical Research Network Center for Hepatic and Digestive Diseases (CIBERehd), Carlos III Health Institute, Madrid, Spain
- Platform for Clinical Research and Clinical Trials IBIMA, Plataforma ISCIII de Investigación Clínica, Madrid, Spain
| | - Raul J Andrade
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain
- Biomedical Research Network Center for Hepatic and Digestive Diseases (CIBERehd), Carlos III Health Institute, Madrid, Spain
| |
Collapse
|
247
|
Memon A, Yeboah-Korang A, Fontana RJ. Advances in the management of idiosyncratic drug-induced liver injury. Clin Liver Dis (Hoboken) 2023; 21:102-106. [PMID: 37936954 PMCID: PMC10627588 DOI: 10.1097/cld.0000000000000052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 04/10/2023] [Indexed: 11/09/2023] Open
Abstract
1_pff0548qKaltura.
Collapse
Affiliation(s)
- Ahmed Memon
- Division of Internal Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Amoah Yeboah-Korang
- Division of Digestive Diseases, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Robert J. Fontana
- Division of Gastroenterology and Hepatology, University of Michigan Health System, Ann Arbor, Michigan, USA
| |
Collapse
|
248
|
Gut microbiota affects sensitivity to immune-mediated isoniazid-induced liver injury. Biomed Pharmacother 2023; 160:114400. [PMID: 36805186 DOI: 10.1016/j.biopha.2023.114400] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/19/2023] Open
Abstract
Isoniazid (INH) is a highly effective single and/or combined first-line anti-tuberculosis (anti-TB) therapy drug, and the hepatotoxicity greatly limits its clinical application. INH-induced liver injury (INH-DILI) is a typical immune-mediated idiosyncratic drug-induced liver injury. Existing mechanisms including genetic variations in drug metabolism and immune responses cannot fully explain the differences in susceptibility and sensitivity to INH-DILI, suggesting that other factors may be involved. Accumulating evidence indicates that the development and severity of immune-mediated liver injury is related to gut microbiota. In this study, INH exposure caused liver damage, immune disregulation and microbiota profile alteration. Depletion of gut microbiota ameliorated INH-DILI, and improved INH-DILI-associated immune disorder and inflammatory response. Moreover, hepatotoxicity of INH was ameliorated by fecal microbiota transplantation (FMT) from INH-treated mice. Notably, Bifidobacterium abundance was significantly associated with transaminase levels. In conclusion, our results suggested that the effect of gut microbiota on INH-DILI was related to immunity, and the difference in INH-DILI sensitivity was related to the structure of gut microbiota. Changes in the structure of gut microbiota by continuous exposure of INH resulted in the tolerance to liver injury, and probiotics such as Bifidobacterium might play an important role in INH-DILI and its "adaptation" phenomenon. This work provides novel evidence for elucidating the underlying mechanism of difference in individual's response to INH-DILI and potential approach for intervening anti-TB drug liver injury by modulating gut microbiota.
Collapse
|
249
|
Wang YF, Ma RX, Zou B, Li J, Yao Y, Li J. Endoplasmic reticulum stress regulates autophagic response that is involved in Saikosaponin a-induced liver cell damage. Toxicol In Vitro 2023; 88:105534. [PMID: 36539104 DOI: 10.1016/j.tiv.2022.105534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 09/28/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022]
Abstract
Saikosaponin a (Ssa) is an active ingredient of the Chinese herbal plant Radix Bupleuri (RB) and has severe hepatotoxicity. However, biomolecular mechanisms involved in Ssa-induced hepatotoxicity are not yet entirely clear. Previous studies reported that Ssd (an isomer of Ssa) as a sarcoplasmic/endoplasmic reticulum calcium ATPase (SERCA) inhibitor can induce autophagy in apoptotic defective cells, leading to autophagy-dependent cell death. Therefore, we speculate that endoplasmic reticulum (ER) stress and autophagy may also play an important role in Ssa-induced hepatocyte death. This study aimed to explore the connection between ER stress and autophagy and Ssa-induced hepatotoxicity. Experiments in vitro showed that the cell viability of L-02 cells in the Ssa treatment group decreased, the level of autophagy marker LC3-II/LC3-I and Beclin1 increased, the level of p62 decreased, the colocalization of autophagosome and lysosome increased, and the cell viability was significantly increased after the application of autophagy inhibitors 3-MA. In addition, SSa can induce ER stress in L-02 cells in vitro. Further studies demonstrated that SSa activated the PERK/eIF2α/ATF4/CHOP pathway, IRE1-TRAF2 pathway, ATF6 pathway, and AMPK/mTOR pathway associated with ER stress. Application of ER stress inhibitors 4-PBA can significantly down-regulate the level of autophagy and improve cell viability. Results of in vivo experiments showed that treatment with 150 and 300 mg/kg Ssa significantly elevated the liver/body weight ratio and caused histological injury in mice liver. Furthermore, Ssa treatment induced significantly downregulated p62 expression but upregulated LC3-II, CHOP, and GRP78 expression in mice livers. Taken together, our results showed that SSa can activate endoplasmic reticulum stress, promote toxic autophagy, and then induce cell death. We revealed an alternative mechanism involving autophagy and ERs, by which Ssa induced hepatotoxicity.
Collapse
Affiliation(s)
- Ye-Feng Wang
- School of Public Health & Management, Ningxia Medical University, Yinchuan 750004, China
| | - Rui-Xia Ma
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Bin Zou
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Jia Li
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Yao Yao
- School of Basic Medical Science, Ningxia Medical University, Yinchuan 750004, China.
| | - Juan Li
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China; Key Laboratory of Modernization of Traditional Chinese Medicine, Ministry of Education, Yinchuan 750004, China.
| |
Collapse
|
250
|
Tong Y, Zhang M, Qi Z, Wu W, Chen J, He F, Han H, Ding P, Wang G, Zhuge Y. Hepatic Venous Occlusion Type of Budd-Chiari Syndrome versus Pyrrolizidine Alkaloid-Induced Hepatic Sinusoidal Obstructive Syndrome: A Multi-Center Retrospective Study. J Pers Med 2023; 13:603. [PMID: 37108988 PMCID: PMC10143067 DOI: 10.3390/jpm13040603] [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: 02/17/2023] [Revised: 03/17/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
(1) Background: Hepatic venous occlusion type of Budd-Chiari syndrome (BCS-HV) and pyrrolizidine alkaloid-induced hepatic sinusoidal obstructive syndrome (PA-HSOS), share similar clinical features, and imaging findings, leading to misdiagnoses; (2) Methods: We retrospectively analyzed 139 patients with BCS-HV and 257 with PA-HSOS admitted to six university-affiliated hospitals. We contrasted the two groups by clinical manifestations, laboratory tests, and imaging features for the most valuable distinguishing indicators.; (3) Results: The mean patient age in BCS-HV is younger than that in PA-HSOS (p < 0.05). In BCS-HV, the prevalence of hepatic vein collateral circulation of hepatic veins, enlarged caudate lobe of the liver, and early liver enhancement nodules were 73.90%, 47.70%, and 8.46%, respectively; none of the PA-HSOS patients exhibited these features (p < 0.05). DUS showed that 86.29% (107/124) of patients with BCS-HV showed occlusion of the hepatic vein, while CT or MRI showed that only 4.55%(5/110) patients had this manifestation (p < 0.001). Collateral circulation of hepatic veins was visible in 70.97% (88/124) of BCS-HV patients on DUS, while only 4.55% (5/110) were visible on CT or MRI (p < 0.001); (4) Conclusions: In addition to an established history of PA-containing plant exposure, local hepatic vein stenosis and the presence of collateral circulation of hepatic veins are the most important differential imaging features of these two diseases. However, these important imaging features may be missed by enhanced CT or MRI, leading to an incorrect diagnosis.
Collapse
Affiliation(s)
- Yaru Tong
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Ming Zhang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Zexue Qi
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Wei Wu
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
| | - Jinjun Chen
- Hepatology Unit, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Fuliang He
- Liver Disease Center, Beijing Friendship Hospital Affiliated to Capital Medical University, Beijing 100050, China
| | - Hao Han
- Department of Ultrasound, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Pengxu Ding
- Department of Intervention, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Guangchuan Wang
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Yuzheng Zhuge
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| |
Collapse
|