1
|
Solar M, Grayck MR, McCarthy WC, Zheng L, Lacayo OA, Sherlock LG, Zhou R, Orlicky DJ, Wright CJ. Absence of IκBβ/NFκB signaling does not attenuate acetaminophen-induced hepatic injury. Anat Rec (Hoboken) 2025; 308:1251-1264. [PMID: 36426684 PMCID: PMC10209348 DOI: 10.1002/ar.25126] [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: 09/13/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 11/27/2022]
Abstract
Acetaminophen (N-acetyl-p-aminophenol [APAP]) toxicity is a common cause of acute liver failure. Innate immune signaling and specifically NFκB activation play a complex role in mediating the hepatic response to toxic APAP exposures. While inflammatory innate immune responses contribute to APAP-induced injury, these same pathways play a role in regeneration and repair. Previous studies have shown that attenuating IκBβ/NFκB signaling downstream of TLR4 activation can limit injury, but whether this pathway contributes to APAP-induced hepatic injury is unknown. We hypothesized that the absence of IκBβ/NFκB signaling in the setting of toxic APAP exposure would attenuate APAP-induced hepatic injury. To test this, we exposed adult male WT and IκBβ-/- mice to APAP (280 mg/kg, IP) and evaluated liver histology at early (2-24 hr) and late (48-72 hr) time points. Furthermore, we interrogated the hepatic expression of NFκB inflammatory (Cxcl1, Tnf, Il1b, Il6, Ptgs2, and Ccl2), anti-inflammatory (Il10, Tnfaip3, and Nfkbia), and Nrf2/antioxidant (Gclc, Hmox, and Nqo1) target genes previously demonstrated to play a role in APAP-induced injury. Conflicting with our hypothesis, we found that hepatic injury was similar in WT and IκBβ-/- mice. Acutely, the induced expression of some target genes was similar in WT and IκBβ-/- mice (Tnfaip3, Nfkbia, and Gclc), while others were either not induced (Cxcl1, Tnf, Ptgs2, and Il10) or significantly attenuated (Ccl2) in IκBβ-/- mice. At later time points, APAP-induced hepatic expression of Il1b, Il6, and Gclc was significantly attenuated in IκBβ-/- mice. Based on these findings, the therapeutic potential of targeting IκBβ/NFκB signaling to treat toxic APAP-induced hepatic injury is likely limited.
Collapse
Affiliation(s)
- Mack Solar
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Maya R. Grayck
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - William C. McCarthy
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Lijun Zheng
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Oscar A. Lacayo
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Laura G. Sherlock
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Ruby Zhou
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - David J. Orlicky
- Dept of Pathology, University of Colorado Anschutz School of Medicine, Aurora, CO
| | - Clyde J. Wright
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| |
Collapse
|
2
|
Yao T, Wu Y, Fu L, Li L. Magnolin ameliorates acetaminophen-induced liver injury in mice via modulating the MAPK pathway and lipid metabolism. Toxicol Appl Pharmacol 2025; 497:117264. [PMID: 39952301 DOI: 10.1016/j.taap.2025.117264] [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: 12/09/2024] [Revised: 02/07/2025] [Accepted: 02/08/2025] [Indexed: 02/17/2025]
Abstract
Acetaminophen (APAP)-induced liver injury (AILI) represents a common yet potentially severe type of drug-induced liver injury with limited available effective therapeutic methods. Magnolin possesses excellent anti-inflammatory and antioxidant properties for treating various diseases. However, its effects against AILI and the fundamental mechanisms still lack comprehensive exploration. This research endeavors to assess magnolin's hepatoprotective properties against AILI. The AILI model was established in male C57BL/6 mice via intraperitoneal injection of 300 mg/kg APAP and in the HepG2 cell line by treating it with 20 mM APAP. The levels of oxidation, liver damage and inflammation were assessed. Transcriptomics and metabolomics were utilized to explore the mechanism underlying magnolin treatment in AILI. We found that 5 mg/kg magnolin effectively mitigated the elevated serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT), along with inflammatory factor (IL-6, and TNF-α) levels in vivo. Meanwhile, magnolin relieved oxidative stress by increasing superoxide dismutase activity and reducing malondialdehyde along with oxidized glutathione/reduced glutathione (GSSG/GSH). 6 μM magnolin increased cell viability and reduced the lipid peroxidation in vitro. Furthermore, 5 mg/kg magnolin altered the expression of 413 genes and the levels of 70 metabolites compared with Control group, which were enriched in lipid metabolism, inflammatory responses, and the MAPK signaling pathway. However, 10 mg/kg magnolin tended to exacerbate liver damage. Overall, 5 mg/kg magnolin effectively protects against AILI by modulating inflammatory responses and the MAPK pathway, whereas 10 mg/kg worsens liver injury, underscoring the need for dose optimization. These findings offer a fresh perspective and novel therapy for AILI.
Collapse
Affiliation(s)
- Ting Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine,79 Qingchun Rd., Hangzhou City 310003, China
| | - Youhe Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine,79 Qingchun Rd., Hangzhou City 310003, China
| | - Liyun Fu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine,79 Qingchun Rd., Hangzhou City 310003, China
| | - Lanjun Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine,79 Qingchun Rd., Hangzhou City 310003, China.
| |
Collapse
|
3
|
Wu Q, Yang D, Liu C, Xu T. Alcohol Plus Additional Risk Factors: Rodent Model of Liver Injury. Semin Liver Dis 2025; 45:81-98. [PMID: 39719149 DOI: 10.1055/a-2490-4278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2024]
Abstract
Alcohol-associated liver disease (ALD), primarily caused by chronic excessive alcohol consumption, is a leading cause of chronic liver disease worldwide. ALD includes alcohol-associated steatotic liver, alcohol-associated hepatitis (AH), fibrosis, cirrhosis, and can even progress to hepatocellular carcinoma (HCC). Existing research indicates that the risk factors of ALD are quite numerous. In addition to drinking patterns, factors such as aldehyde dehydrogenase 2 (ALDH2) deficiency, smoking, medication administration, high-fat diet (HFD), hepatitis virus infection, and disruption of circadian rhythms can also increase susceptibility to ALD. However, there is limited understanding regarding the exacerbation of liver injury by alcohol plus additional risk factors. This review presents rodent models of EtOH + "X," which simulate the synergistic effects of alcohol and additional risk factors in causing liver injury. These models offer a further exploration of the interactions between alcohol and additional risk factors, advancing the simulation of human ALD and providing a more reliable platform for studying disease mechanisms and exploring therapeutic interventions. We summarize the modeling methods, relevant indicators of liver injury, and focus on the targets of the synergistic effects as well as the associated mechanisms.
Collapse
Affiliation(s)
- Qixiang Wu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, P.R. China
- Institute for Liver Diseases of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Dashuai Yang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, P.R. China
- Institute for Liver Diseases of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Chixiang Liu
- Department of Blood Transfusion, Southern Medical University, Nanfang Hospital, Guangzhou, P.R. China
- School of Laboratory and Biotechnology, Institute of Antibody Engineering, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Tao Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, P.R. China
- Institute for Liver Diseases of Anhui Medical University, Hefei, Anhui, P.R. China
| |
Collapse
|
4
|
Tian W, Li Y, Liu F, Liu H, Li C, Bao L, Liang X. Strychni Semen and two alkaloidal components cause apoptosis in HK-2 cells through TRADD-MAPK/NF-κB pathway. Toxicon 2025; 256:108224. [PMID: 39798900 DOI: 10.1016/j.toxicon.2024.108224] [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: 10/12/2024] [Revised: 12/16/2024] [Accepted: 12/30/2024] [Indexed: 01/15/2025]
Abstract
Strychni Semen is the dried ripe seeds of the plant Strychnos nux-vomica L, and has great medicinal value and developmental potential.However, Strychni Semen is severely toxic, with adverse effects on the central nervous system, urinary system, and other organ systems, and severe cases can be life-threatening. The present study was to reveal the mechanism of nephrotoxicity induced by Strychni Semen and its alkaloid components using experiments. HK-2 cells were randomly divided into control, experimental, and inhibitor groups. The experimental group was divided into Strychni Semen (SS, 10 mg/mL), brucine (B, 8 μg/mL) and strychnine (S, 4 μg/mL) groups,and the inhibitor group was treated with 1 μm/L Apostatin-1. To detect the effects of each group of drugs on the expression of inflammatory cytokines, KIM-1 and TRADD downstream pathway-related proteins. Network pharmacology predicted that nephrotoxicity caused by Strychni Semen may be related to MAPK. Cell experiments showed that Strychni Semen and its alkaloids could induce the activation of the JNK and p38 pathways in the NF-κB and MAPK pathways, upregulate the activation and expression of caspase-3, promote the apoptosis of HK-2 cells, and enhance the production of the cytokines IL-6, IL-1β, and TNF-α and KIM-1. Apostatin-1 antagonises the apoptosis of HK-2 cells induced by Strychni Semen and its alkaloids and reduces the production of the above-mentioned cytokines. The results showed that Strychni Semen and its alkaloids can induce apoptosis of HK-2 cells by activating TRADD-mediated MAPK and NF-κB pathways, showing cytotoxicity to HK-2 cells. Thus, inhibiting TRADD can reduce apoptosis.
Collapse
Affiliation(s)
- Wenyi Tian
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Yuling Li
- Shandong Wendeng Osteopathic Hospital, 1 Fengshan Road, Wendeng District, Weihai, China.
| | - Fengzhi Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Hui Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Chen Li
- Department of Education, Shandong Provincial Third Hospital, Jinan, 250031, Shandong, China.
| | - Lin Bao
- The 960th Hospital of the Joint Logistic Support Force of the people's liberation Army, Jinan, 250031, China.
| | - Xiaodong Liang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
5
|
Zhang Y, Shen Z, Mao Z, Huang D, Lou C, Fang L. VPO1 Promotes Programmed Necrosis of Cardiomyocytes in Rats with Chronic Heart Failure by Upregulating CYLD. FRONT BIOSCI-LANDMRK 2024; 29:425. [PMID: 39735991 DOI: 10.31083/j.fbl2912425] [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: 06/25/2024] [Revised: 10/10/2024] [Accepted: 10/22/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND Chronic heart failure (CHF) is a serious cardiovascular condition. Vascular peroxidase 1 (VPO1) is associated with various cardiovascular diseases, yet its role in CHF remains unclear. This research aims to explore the involvement of VPO1 in CHF. METHODS CHF was induced in rats using adriamycin, and the expression levels of VPO1 and cylindromatosis (CYLD) were assessed. In parallel, the effects of VPO1 on programmed necrosis in H9c2 cells were evaluated through cell viability assays, lactate dehydrogenase (LDH) level measurements, and analysis of receptor-interacting protein kinase 1/receptor-interacting protein kinase 3/mixed lineage kinase domain-like protein (RIPK1/RIPK3/MLKL) pathway-related proteins. The impact of CYLD on RIPK1 protein stability and ubiquitination was also investigated, along with the interaction between VPO1 and CYLD. Additionally, cardiac structure and function were assessed using echocardiography, Hematoxylin-eosin (HE) staining, Masson staining, and measurements of myocardial injury-related factors, including N-terminal prohormone of brain natriuretic peptide (NT-proBNP), Aspartate aminotransferase (AST), LDH, and creatine kinase-myocardial band (CK-MB). RESULTS VPO1 expression was upregulated in CHF rats and in H9c2 cells treated with adriamycin. In cellular experiments, VPO1 knockdown improved cell viability, inhibited necrosis and the expression of proteins associated with the RIPK1/RIPK3/MLKL pathway. Mechanistically, VPO1 promoted cardiomyocyte programmed necrosis by interacting with the deubiquitinating enzyme CYLD, which enhanced RIPK1 ubiquitination and degradation, leading to activation of the RIPK1/RIPK3/MLKL signaling pathway. At animal level, overexpression of CYLD counteracted the cardiac failure, cardiac hypertrophy, myocardial injury, myocardial fibrosis, and tissue necrosis caused by VPO1 knockdown. CONCLUSIONS VPO1 exacerbates cardiomyocyte programmed necrosis in CHF rats by upregulating CYLD, which activates the RIPK1/RIPK3/MLKL signaling pathway. Thus, VPO1 may represent a potential therapeutic target for CHF.
Collapse
Affiliation(s)
- Yinzhuang Zhang
- Department of Cardiovascular Medicine, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, 410008 Changsha, Hunan, China
| | - Zhijie Shen
- Department of Cardiovascular Medicine, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, 410008 Changsha, Hunan, China
| | - Zhuoni Mao
- Department of Cardiovascular Medicine, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, 410008 Changsha, Hunan, China
| | - Dan Huang
- Department of Cardiovascular Medicine, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, 410008 Changsha, Hunan, China
| | - Chengyu Lou
- Department of Cardiovascular Medicine, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, 410008 Changsha, Hunan, China
| | - Li Fang
- Department of Cardiovascular Medicine, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, 410008 Changsha, Hunan, China
| |
Collapse
|
6
|
Deng L, Wei SL, Wang L, Huang JQ. Feruloylated Oligosaccharides Prevented Influenza-Induced Lung Inflammation via the RIG-I/MAVS/TRAF3 Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:9782-9794. [PMID: 38597360 DOI: 10.1021/acs.jafc.3c09390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Uncontrolled inflammation contributes significantly to the mortality in acute respiratory infections. Our previous research has demonstrated that maize bran feruloylated oligosaccharides (FOs) possess notable anti-inflammatory properties linked to the NF-kB pathway regulation. In this study, we clarified that the oral administration of FOs moderately inhibited H1N1 virus infection and reduced lung inflammation in influenza-infected mice by decreasing a wide spectrum of cytokines (IFN-α, IFN-β, IL-6, IL-10, and IL-23) in the lungs. The mechanism involves FOs suppressing the transduction of the RIG-I/MAVS/TRAF3 signaling pathway, subsequently lowering the expression of NF-κB. In silico analysis suggests that FOs have a greater binding affinity for the RIG-I/MAVS signaling complex. This indicates that FOs have potential as promising targets for immune modulation. Moreover, in MAVS knockout mice, we confirmed that the anti-inflammatory function of FOs against influenza depends on MAVS. Comprehensive analysis using 16S rRNA gene sequencing and metabolite profiling techniques showed that FOs have the potential to restore immunity by modulating the gut microbiota. In conclusion, our study demonstrates that FOs are effective anti-inflammatory phytochemicals in inhibiting lung inflammation caused by influenza. This suggests that FOs could serve as a potential nutritional strategy for preventing the H1N1 virus infection and associated lung inflammation.
Collapse
Affiliation(s)
- Li Deng
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Shu-Lei Wei
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Lu Wang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Jun-Qing Huang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| |
Collapse
|
7
|
Xu Y, Xu H, Ling T, Cui Y, Zhang J, Mu X, Zhou D, Zhao T, Li Y, Su Z, You Q. Inhibitor of nuclear factor kappa B kinase subunit epsilon regulates murine acetaminophen toxicity via RIPK1/JNK. Cell Biol Toxicol 2023; 39:2709-2724. [PMID: 36757501 DOI: 10.1007/s10565-023-09796-8] [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: 09/20/2022] [Accepted: 01/31/2023] [Indexed: 02/10/2023]
Abstract
Drug-induced liver injury (DILI) still poses a major clinical challenge and is a leading cause of acute liver failure. Inhibitor of nuclear factor kappa B kinase subunit epsilon (IKBKE) is essential for inflammation and metabolic disorders. However, it is unclear how IKBKE regulates cellular damage in acetaminophen (APAP)-induced acute liver injury. Here, we found that the deficiency of IKBKE markedly aggravated APAP-induced acute liver injury by targeting RIPK1. We showed that APAP-treated IKBKE-deficient mice exhibited severer liver injury, worse mitochondrial integrity, and enhanced glutathione depletion than wild-type mice. IKBKE deficiency may directly upregulate the expression of total RIPK1 and the cleaved RIPK1, resulting in sustained JNK activation and increased translocation of RIPK1/JNK to mitochondria. Moreover, deficiency of IKBKE enhanced the expression of pro-inflammatory factors and inflammatory cell infiltration in the liver, especially neutrophils and monocytes. Inhibition of RIPK1 activity by necrostatin-1 significantly reduced APAP-induced liver damage. Thus, we have revealed a negative regulatory function of IKBKE, which acts as an RIPK1/JNK regulator to mediate APAP-induced hepatotoxicity. Targeting IKBKE/RIPK1 may serve as a potential therapeutic strategy for acute or chronic liver injury.
Collapse
Affiliation(s)
- Yujie Xu
- Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou, China
| | - Haozhe Xu
- Department of Biotherapy, Medical Center for Digestive Diseases, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Tao Ling
- Department of Biotherapy, Medical Center for Digestive Diseases, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Yachao Cui
- Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou, China
| | - Junwei Zhang
- Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou, China
| | - Xianmin Mu
- Department of Biotherapy, Medical Center for Digestive Diseases, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Desheng Zhou
- Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou, China
| | - Ting Zhao
- Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou, China
| | - Yingchang Li
- Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou, China
| | - Zhongping Su
- Department of Geriatric Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine Tumor, Nanjing Medical University, Nanjing, China.
| | - Qiang You
- Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou, China.
- Department of Biotherapy, Medical Center for Digestive Diseases, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
8
|
Yiew NKH, Vazquez JH, Martino MR, Kennon-McGill S, Price JR, Allard FD, Yee EU, Layman AJ, James LP, McCommis KS, Finck BN, McGill MR. Hepatic pyruvate and alanine metabolism are critical and complementary for maintenance of antioxidant capacity and resistance to oxidative insult. Mol Metab 2023; 77:101808. [PMID: 37716594 PMCID: PMC10561123 DOI: 10.1016/j.molmet.2023.101808] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/16/2023] [Accepted: 09/12/2023] [Indexed: 09/18/2023] Open
Abstract
OBJECTIVE Mitochondrial pyruvate is a critical intermediary metabolite in gluconeogenesis, lipogenesis, and NADH production. As a result, the mitochondrial pyruvate carrier (MPC) complex has emerged as a promising therapeutic target in metabolic diseases. Clinical trials are currently underway. However, recent in vitro data indicate that MPC inhibition diverts glutamine/glutamate away from glutathione synthesis and toward glutaminolysis to compensate for loss of pyruvate oxidation, possibly sensitizing cells to oxidative insult. Here, we explored this in vivo using the clinically relevant acetaminophen (APAP) overdose model of acute liver injury, which is driven by oxidative stress. METHODS We used pharmacological and genetic approaches to inhibit MPC2 and alanine aminotransferase 2 (ALT2), individually and concomitantly, in mice and cell culture models and determined the effects on APAP hepatotoxicity. RESULTS We found that MPC inhibition sensitizes the liver to APAP-induced injury in vivo only with concomitant loss of alanine aminotransferase 2 (ALT2). Pharmacological and genetic manipulation of neither MPC2 nor ALT2 alone affected APAP toxicity, but liver-specific double knockout (DKO) significantly worsened APAP-induced liver damage. Further investigation indicated that DKO impaired glutathione synthesis and increased urea cycle flux, consistent with increased glutaminolysis, and these results were reproducible in vitro. Finally, induction of ALT2 and post-treatment with dichloroacetate both reduced APAP-induced liver injury, suggesting new therapeutic avenues. CONCLUSIONS Increased susceptibility to APAP toxicity requires loss of both the MPC and ALT2 in vivo, indicating that MPC inhibition alone is insufficient to disrupt redox balance. Furthermore, the results from ALT2 induction and dichloroacetate in the APAP model suggest new metabolic approaches to the treatment of liver damage.
Collapse
Affiliation(s)
- Nicole K H Yiew
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Joel H Vazquez
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA; Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Michael R Martino
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Stefanie Kennon-McGill
- Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jake R Price
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA; Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Felicia D Allard
- Department of Pathology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Eric U Yee
- Department of Pathology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Alexander J Layman
- Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Laura P James
- Department of Pediatrics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Kyle S McCommis
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Brian N Finck
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Mitchell R McGill
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA; Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Department of Pathology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
9
|
Zheng J, Yang N, Wan Y, Cheng W, Zhang G, Yu S, Yang B, Liu X, Chen X, Ding X, Wu L, Yu X. Celastrol-loaded biomimetic nanodrug ameliorates APAP-induced liver injury through modulating macrophage polarization. J Mol Med (Berl) 2023:10.1007/s00109-023-02321-8. [PMID: 37129620 DOI: 10.1007/s00109-023-02321-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 05/03/2023]
Abstract
Drug-induced liver injury (DILI) is a major concern in clinical treatment as well as postmarketing surveillance, showing an urgent requirement for the development of protective medications. Celastrol (Cel), a highly active natural product extracted from the roots of Tripterygium wilfordii, has a potential liver protective activity due to its antioxidant and anti-inflammatory effects. However, the further application of Cel to DILI remains a challenge because of its short half-life, low solubility, and toxic side effects. Herein, we developed a Cel-loaded biomimetic nanodrug based on erythrocyte membrane vesicles (EMV) for protecting the liver from acetaminophen (APAP)-induced liver injury. The Cel-loaded EMV (C-EMV) with lower cytotoxicity had a well-sustained release effect and exhibited excellent ability for liver accumulation under physiological and pathological conditions. By suppressing the inflammatory response of pro-inflammatory macrophage M1 polarization while stimulating anti-inflammatory macrophage M2 polarization, C-EMV could significantly alleviate the primary pathological manifestations related to liver injury, including aberrant elevation of biochemical indicators, histopathological alterations, neutrophil infiltration as well as hepatocyte DNA fragmentation. The macrophage depletion experiment further demonstrated that the protective effect of C-EMV on APAP-induced liver injury appeared to be dependent on hepatic macrophages. Therefore, C-EMV as a biomimetic nanodrug exhibits great potential for attenuating the progress of DILI, providing a new approach to protecting the liver from DILI as well as other liver inflammatory diseases through a targeted nanodelivery system. KEY MESSAGES: EMV biomimetic nanocarrier has good monodispersity and sustained-release property. EMV biomimetic nanocarrier displays excellent liver-targeting capability under physiological and pathological conditions. C-EMV biomimetic nanodrug with lower cytotoxicity regulates macrophage polarization in vitro and in vivo. C-EMV biomimetic nanodrug can significantly alleviate APAP-induced liver injury. The protective effect of C-EMV on APAP-induced liver injury is dependent on hepatic macrophages.
Collapse
Affiliation(s)
- Jing Zheng
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, China Three Gorges University, Yichang, China
- Medical College, China Three Gorges University, Yichang, China
- The People's Hospital of China Three Gorges University, Yichang, China
| | - Ni Yang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, China Three Gorges University, Yichang, China
- Medical College, China Three Gorges University, Yichang, China
| | - Yingying Wan
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, China Three Gorges University, Yichang, China
- Medical College, China Three Gorges University, Yichang, China
| | - Wenjing Cheng
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, China Three Gorges University, Yichang, China
- Medical College, China Three Gorges University, Yichang, China
| | - Gan Zhang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, China Three Gorges University, Yichang, China
- Medical College, China Three Gorges University, Yichang, China
| | - Shi Yu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, China Three Gorges University, Yichang, China
- Medical College, China Three Gorges University, Yichang, China
| | - Baoye Yang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, China Three Gorges University, Yichang, China
- Medical College, China Three Gorges University, Yichang, China
| | - Xinyu Liu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, China Three Gorges University, Yichang, China
- Medical College, China Three Gorges University, Yichang, China
| | - Xingyan Chen
- Tong Ji Hospital, Tongji Medical College of HUST, Wuhan, China
| | - Xueliang Ding
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- Medical College, China Three Gorges University, Yichang, China
- Department of Clinical Laboratory, Affiliated Renhe Hospital of China Three Gorges University, Yichang, China
| | - Ling Wu
- The People's Hospital of China Three Gorges University, Yichang, China.
| | - Xiang Yu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China.
- Institute of Infection and Inflammation, China Three Gorges University, Yichang, China.
- Medical College, China Three Gorges University, Yichang, China.
| |
Collapse
|
10
|
Kim HY, Yoon HS, Heo AJ, Jung EJ, Ji CH, Mun SR, Lee MJ, Kwon YT, Park JW. Mitophagy and endoplasmic reticulum-phagy accelerated by a p62 ZZ ligand alleviates paracetamol-induced hepatotoxicity. Br J Pharmacol 2022; 180:1247-1266. [PMID: 36479690 DOI: 10.1111/bph.16004] [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: 07/05/2022] [Revised: 10/31/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Paracetamol (acetaminophen)-induced hepatotoxicity is the leading cause of drug-induced liver injury worldwide. Autophagy is a degradative process by which various cargoes are collected by the autophagic receptors such as p62/SQSTM1/Sequestosome-1 for lysosomal degradation. Here, we investigated the protective role of p62-dependent autophagy in paracetamol-induced liver injury. EXPERIMENTAL APPROACH Paracetamol-induced hepatotoxicity was induced by a single i.p. injection of paracetamol (500 mg·kg-1 ) in C57/BL6 male mice. YTK-2205 (20 mg·kg-1 ), a p62 agonist targeting ZZ domain, was co- or post-administered with paracetamol. Western blotting and immunocytochemistry were performed to explore the mechanism. KEY RESULTS N-terminal arginylation of the molecular chaperone calreticulin retro-translocated from the endoplasmic reticulum (ER) was induced in the livers undergoing paracetamol-induced hepatotoxicity, and YTK-2205 exhibited notable therapeutic efficacy in acute hepatotoxicity as assessed by the levels of serum alanine aminotransferase and hepatic necrosis. This efficacy was significantly attributed to accelerated degradation of ubiquitin (Ub) conjugates as well as damaged mitochondria (mitophagy) and endoplasmic reticulum (ER-phagy). In primary murine hepatocytes treated with paracetamol, YTK-2205 induced the co-localization of p62+ LC3+ phagophores to the sites of mitophagy and ER-phagy. A similar activity of YTK-2205 was observed with N-acetyl-p-benzoquinone imine, a putative toxic metabolite of paracetamol in Hep3B cells. CONCLUSION AND IMPLICATIONS Our results elucidated that p62-dependent autophagy plays a key role in the removal of cytotoxic materials such as damaged mitochondria in paracetamol-induced hepatotoxicity. Small molecule ligands to p62 may be developed into drugs to treat this pathological condition.
Collapse
Affiliation(s)
- Hee-Yeon Kim
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Hee-Soo Yoon
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Ah Jung Heo
- Cellular Degradation Biology Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Eui Jung Jung
- Cellular Degradation Biology Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Chang Hoon Ji
- Cellular Degradation Biology Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea.,AUTOTAC Bio Inc., 254, Changgyeonggung-ro, Jongno-gu, Seoul, Republic of Korea
| | - Su Ran Mun
- Cellular Degradation Biology Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Min Ju Lee
- Cellular Degradation Biology Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Yong Tae Kwon
- Cellular Degradation Biology Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea.,AUTOTAC Bio Inc., 254, Changgyeonggung-ro, Jongno-gu, Seoul, Republic of Korea.,Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea.,SNU Dementia Research Center, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Joo-Won Park
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
11
|
Basta DW, Vong M, Beshimova A, Nakamura BN, Rusu I, Kattah MG, Shao L. A20 Restricts NOS2 Expression and Intestinal Tumorigenesis in a Mouse Model of Colitis-Associated Cancer. GASTRO HEP ADVANCES 2022; 2:96-107. [PMID: 36636264 PMCID: PMC9833806 DOI: 10.1016/j.gastha.2022.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
BACKGROUND AND AIMS Colon cancer can occur sporadically or in the setting of chronic inflammation, such as in patients with inflammatory bowel disease. We previously showed that A20, a critical negative regulator of tumor necrosis factor signal transduction, could regulate sporadic colon cancer development. In this report, we investigate whether A20 also acts as a tumor suppressor in a model of colitis-associated cancer. METHODS Colitis and colitis-associated tumors were induced in wild-type and A20 intestinal epithelial cell-specific knockout (A20dIEC) mice using dextran sodium sulfate and azoxymethane. Clinicopathologic markers of inflammation were assessed in conjunction with colonic tumor burden. Gene expression analyses and immunohistochemistry were performed on colonic tissue and intestinal enteroids. Nitric oxide (NO) production and activity were assessed in whole colonic lysates and mouse embryonic fibroblasts. RESULTS A20dIEC mice develop larger tumors after treatment with dextran sodium sulfate and azoxymethane than wild-type mice. In addition to elevated markers of inflammation, A20dIEC mice have significantly enhanced expression of inducible nitric oxide synthase (iNOS), a well-known driver of neoplasia. Enhanced iNOS expression is associated with the formation of reactive nitrogen species and DNA damage. Loss of A20 also enhances NO-dependent cell death directly. CONCLUSION Mechanistically, we propose that A20 normally restricts tumor necrosis factor-induced nuclear factor kappa B-dependent production of iNOS in intestinal epithelial cells, thereby protecting against colitis-associated tumorigenesis. We also propose that A20 plays a direct role in regulating NO-dependent cell death.
Collapse
Affiliation(s)
- David W Basta
- Division of Gastroenterology and Liver Disease, Department of Medicine, University of Southern California, Keck School of Medicine, Los Angeles, California
| | - Mandy Vong
- Division of Gastroenterology and Liver Disease, Department of Medicine, University of Southern California, Keck School of Medicine, Los Angeles, California
| | - Adolat Beshimova
- Division of Gastroenterology and Liver Disease, Department of Medicine, University of Southern California, Keck School of Medicine, Los Angeles, California
| | - Brooke N Nakamura
- Division of Gastroenterology and Liver Disease, Department of Medicine, University of Southern California, Keck School of Medicine, Los Angeles, California
| | - Iulia Rusu
- Division of Gastroenterology and Liver Disease, Department of Medicine, University of Southern California, Keck School of Medicine, Los Angeles, California
| | - Michael G Kattah
- Division of Gastroenterology and Liver Disease, Department of Medicine, University of Southern California, Keck School of Medicine, Los Angeles, California
| | - Ling Shao
- Division of Gastroenterology and Liver Disease, Department of Medicine, University of Southern California, Keck School of Medicine, Los Angeles, California
| |
Collapse
|
12
|
Bai W, Huo S, Li J, Shao J. Advances in the Study of the Ubiquitin-Editing Enzyme A20. Front Pharmacol 2022; 13:845262. [PMID: 35592427 PMCID: PMC9110840 DOI: 10.3389/fphar.2022.845262] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/28/2022] [Indexed: 11/30/2022] Open
Abstract
Ubiquitin modification is a common post-translational protein modification and an important mechanism whereby the body regulates protein levels and functions. As a common enzyme associated with ubiquitin modification, the ubiquitin-editing enzyme A20 may be closely associated with the development of numerous pathological processes through its different structural domains. The aim of this paper is to provide an overview of the following: advances in ubiquitination research, the structure and function of A20, and the relationships between A20 and immune inflammatory response, apoptosis, necroptosis, pyroptosis, and autophagy.
Collapse
Affiliation(s)
- Wenya Bai
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Siying Huo
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Junjie Li
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jianlin Shao
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
13
|
Chen H, McKeen T, Chao X, Chen A, Deng F, Jaeschke H, Ding WX, Ni HM. The role of MLKL in Hepatic Ischemia-Reperfusion Injury of Alcoholic Steatotic Livers. Int J Biol Sci 2022; 18:1096-1106. [PMID: 35173541 PMCID: PMC8771841 DOI: 10.7150/ijbs.67533] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/14/2021] [Indexed: 12/15/2022] Open
Abstract
Alcohol-related liver disease (ALD) and non-alcoholic fatty liver disease (NAFLD) are the primary causes of chronic liver disease in western countries. Liver transplantation is currently one of the most efficient approaches to save patients with liver failure, which is often associated with hepatic ischemia-reperfusion (IR) injury. IR injury is exacerbated by hepatic steatosis, yet the mechanism remains elusive. Necroptosis is a form of regulated cell death mediated by receptor-interacting protein kinase 1 (RIP1), RIP3 and mixed lineage kinase domain-like (MLKL) protein, which has been implicated in the pathogenesis of ALD and NAFLD. Though necroptosis plays an important role in IR injury of high fat diet - induced steatotic livers, the role of necroptosis in IR injury of ethanol - induced steototic livers has not been investigated. In the present study, we used chronic plus binge alcohol (Gao-binge) feeding followed by IR surgery to investigate IR liver injury with ethanol-associated steatosis. We found that the levels of key necroptotic proteins MLKL and RIP3 increased in alcohol-fed mouse livers. Moreover, we observed increased liver injury after IR in control diet-fed mice, which was further exacerbated by alcohol feeding based on serum alanine aminotransferase (ALT) levels and TUNEL staining of necrotic cells. Hepatic neutrophil infiltration also increased in alcohol-fed mice after IR surgery. However, deletion of Mlkl did not protect against IR liver injury in alcohol-fed mice compared with matched wild-type mice. In conclusion, alcoholic steatosis promotes IR injury, which seems to be independent of MLKL-mediated necroptosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Hong-Min Ni
- ✉ Corresponding author: Hong-Min Ni, MD., Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, MS 1018, 3901 Rainbow Blvd., Kansas City, Kansas 66160, Phone: 913-588-8999;
| |
Collapse
|
14
|
Hameed H, Farooq M, Piquet-Pellorce C, Hamon A, Samson M, Le Seyec J. Questioning the RIPK1 kinase activity involvement in acetaminophen-induced hepatotoxicity in mouse. Free Radic Biol Med 2022; 178:243-245. [PMID: 34879229 DOI: 10.1016/j.freeradbiomed.2021.11.042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/18/2021] [Accepted: 11/30/2021] [Indexed: 12/14/2022]
Affiliation(s)
- Huma Hameed
- Univ Rennes, Inserm, EHESP, IRSET (Institut de recherche en santé, environnement et travail) - UMR_S, 1085, Rennes, France
| | - Muhammad Farooq
- Univ Rennes, Inserm, EHESP, IRSET (Institut de Recherche en santé, environnement et travail) - UMR_S, 1085, Rennes, France; Department of Clinical Sciences, College of Veterinary and Animal Sciences, Jhang, Pakistan
| | - Claire Piquet-Pellorce
- Univ Rennes, Inserm, EHESP, IRSET (Institut de Recherche en santé, environnement et travail) - UMR_S, 1085, Rennes, France
| | - Annaïg Hamon
- Univ Rennes, Inserm, EHESP, IRSET (Institut de Recherche en santé, environnement et travail) - UMR_S, 1085, Rennes, France
| | - Michel Samson
- Univ Rennes, Inserm, EHESP, IRSET (Institut de Recherche en santé, environnement et travail) - UMR_S, 1085, Rennes, France.
| | - Jacques Le Seyec
- Univ Rennes, Inserm, EHESP, IRSET (Institut de Recherche en santé, environnement et travail) - UMR_S, 1085, Rennes, France
| |
Collapse
|
15
|
Yi Y, Zhang W, Tao L, Shao Q, Xu Q, Chen Y, Zhang H, Zhang J, Weng D. RIP1 kinase inactivation protects against acetaminophen-induced acute liver injury in mice. Free Radic Biol Med 2021; 174:57-65. [PMID: 34324981 DOI: 10.1016/j.freeradbiomed.2021.07.034] [Citation(s) in RCA: 6] [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: 05/25/2021] [Revised: 07/20/2021] [Accepted: 07/25/2021] [Indexed: 02/05/2023]
Abstract
Many studies have investigated the role of receptor-interacting protein 1 (RIP1) kinase in acetaminophen (APAP) overdose-induced acute liver injury. However, the results were not consistent and there still remain controversies. Importantly, in these previous studies, the usage of DMSO to dissolve the RIP1 kinase inhibitor Nec-1, resulted in misleading conclusion. Our study aimed to determine the role of RIP1 kinase in APAP-induced liver injury, via genetically or pharmaceutically inhibition of RIP1 kinase activity. Our results indicated that APAP-induced liver injury was significantly attenuated in RIP1 kinase-dead (Rip1K45A/K45A) mice compared to WT control. High dosage of APAP-induced mortality was also rescued by RIP1 kinase inactivation. In agreement, RIP1 kinase inhibitor, Nec-1 which was formulated with PEG400, could efficiently alleviate APAP-induced hepatotoxicity. For the underlying mechanism, our results suggested that RIP1 kinase inactivation did not influence the hepatic GSH depletion, but significantly reduced the hepatic cell death and inflammation induced by APAP treatment. Using bone marrow transplantation model, we also demonstrated that it was RIP1 kinase activity in tissue-resident hepatic cells other than hematopoietic-derived cells mainly responsible for APAP-induced liver injury. Our study confirmed the important role of RIP1 kinase activity in APAP-induced acute liver failure.
Collapse
Affiliation(s)
- Yuguo Yi
- Center for Molecular Metabolism, Nanjing University of Science & Technology, 200 Xiaolingwei Street, Nanjing, 210094, China
| | - Weigao Zhang
- Center for Molecular Metabolism, Nanjing University of Science & Technology, 200 Xiaolingwei Street, Nanjing, 210094, China
| | - Liang Tao
- Center for Molecular Metabolism, Nanjing University of Science & Technology, 200 Xiaolingwei Street, Nanjing, 210094, China
| | - Qianchao Shao
- Center for Molecular Metabolism, Nanjing University of Science & Technology, 200 Xiaolingwei Street, Nanjing, 210094, China
| | - Qian Xu
- Center for Molecular Metabolism, Nanjing University of Science & Technology, 200 Xiaolingwei Street, Nanjing, 210094, China
| | - Yuxin Chen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Haibing Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 100864, China
| | - Jianfa Zhang
- Center for Molecular Metabolism, Nanjing University of Science & Technology, 200 Xiaolingwei Street, Nanjing, 210094, China
| | - Dan Weng
- Center for Molecular Metabolism, Nanjing University of Science & Technology, 200 Xiaolingwei Street, Nanjing, 210094, China.
| |
Collapse
|
16
|
Li T, Zhang J, Wang PJ, Zhang ZW, Huang JQ. Selenoproteins Protect Against Avian Liver Necrosis by Metabolizing Peroxides and Regulating Receptor Interacting Serine Threonine Kinase 1/Receptor Interacting Serine Threonine Kinase 3/Mixed Lineage Kinase Domain-Like and Mitogen-Activated Protein Kinase Signaling. Front Physiol 2021; 12:696256. [PMID: 34456747 PMCID: PMC8397447 DOI: 10.3389/fphys.2021.696256] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/12/2021] [Indexed: 12/17/2022] Open
Abstract
Liver necroptosis of chicks is induced by selenium (Se)/vitamin E (VE) deficiencies and may be associated with oxidative cell damage. To reveal the underlying mechanisms of liver necrosis, a pool of the corn-soy basal diet (10 μg Se/kg; no VE added), a basal diet plus all-rac-α-tocopheryl acetate (50 mg/kg), Se (sodium selenite at 0.3 mg/kg), or both of these nutrients were provided to day-old broiler chicks (n = 40/group) for 6 weeks. High incidences of liver necrosis (30%) of chicks were induced by -SE-VE, starting at day 16. The Se concentration in liver and glutathione peroxidase (GPX) activity were decreased (P < 0.05) by dietary Se deficiency. Meanwhile, Se deficiency elevated malondialdehyde content and decreased superoxide dismutase (SOD) activity in the liver at weeks 2 and 4. Chicks fed with the two Se-deficient diets showed lower (P < 0.05) hepatic mRNA expression of Gpx1, Gpx3, Gpx4, Selenof, Selenoh, Selenok, Selenom, Selenon, Selenoo, Selenop, Selenot, Selenou, Selenow, and Dio1 than those fed with the two Se-supplemented diets. Dietary Se deficiency had elevated (P < 0.05) the expression of SELENOP, but decreased the downregulation (P < 0.05) of GPX1, GPX4, SELENON, and SELENOW in the liver of chicks at two time points. Meanwhile, dietary Se deficiency upregulated (P < 0.05) the abundance of hepatic proteins of p38 mitogen-activated protein kinase, phospho-p38 mitogen-activated protein kinase, c-Jun N-terminal kinase, phospho-c-Jun N-terminal kinase, extracellular signal-regulated kinase, phospho-mitogen-activated protein kinase, receptor-interacting serine-threonine kinase 1 (RIPK1), receptor-interacting serine-threonine kinase 3 (RIPK3), and mixed lineage kinase domain-like (MLKL) at two time points. In conclusion, our data confirmed the differential regulation of dietary Se deficiency on several key selenoproteins, the RIPK1/RIPK3/MLKL, and mitogen-activated protein kinase signaling pathway in chicks and identified new molecular clues for understanding the etiology of nutritional liver necrosis.
Collapse
Affiliation(s)
- Tong Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Jing Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, China Agricultural University, Beijing, China
- Administrative Engineering College, Xu Zhou University of Technology, Xuzhou, China
| | - Peng-Jie Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, China Agricultural University, Beijing, China
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Zi-Wei Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, China Agricultural University, Beijing, China
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Jia-Qiang Huang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, China Agricultural University, Beijing, China
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
| |
Collapse
|
17
|
Kim W, Tokuda H, Tanabe K, Yamaguchi S, Hioki T, Tachi J, Matsushima-Nishiwaki R, Kozawa O, Iida H. Acetaminophen reduces osteoprotegerin synthesis stimulated by PGE 2 and PGF 2α in osteoblasts: attenuation of SAPK/JNK but not p38 MAPK or p44/p42 MAPK. Biomed Res 2021; 42:77-84. [PMID: 33840687 DOI: 10.2220/biomedres.42.77] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Acetaminophen is one of the most widely used analgesic and antipyretic medicines, whose long-period use has reportedly been associated with an increased risk of bone fracture. However, the mechanism underlying this undesired effect remains to be investigated. The homeostatic control of bone tissue depends on the interaction between osteoblasts and osteoclasts. Osteoprotegerin produced by osteoblasts is known to play an essential role in suppressing osteoclast induction. We have previously reported that prostaglandin (PG) E2 and PGF2α induce osteoprotegerin synthesis through p38 mitogen-activated protein kinase (MAPK), p44/p42 MAPK and stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) in osteoblast-like MC3T3-E1 cells. In the present study, we investigated the effects of acetaminophen on the osteoprotegerin synthesis induced by PGE2 and PGF2α in MC3T3-E1 cells. Acetaminophen significantly suppressed the osteoprotegerin release stimulated by PGE2 and PGF2α. The PGE2-induced expression of osteoprotegerin mRNA was also reduced by acetaminophen. Acetaminophen markedly downregulated the phosphorylation of SAPK/JNK stimulated by PGE2 and PGF2α, but not those of p38 MAPK or p44/p42 MAPK. SP600125, an inhibitor of SAPK/JNK, suppressed the levels of PGE2- and PGF2α-upregulated osteoprotegerin mRNA expression. Taken together, these results strongly suggest that acetaminophen reduces the PGE2- and PGF2α-stimulated synthesis of osteoprotegerin in osteoblasts, and that the suppressive effect is exerted via attenuation of SAPK/JNK. These findings provide a molecular basis for the possible effect of acetaminophen on bone tissue metabolism.
Collapse
Affiliation(s)
- Woo Kim
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine.,Department of Pharmacology, Gifu University Graduate School of Medicine
| | - Haruhiko Tokuda
- Department of Pharmacology, Gifu University Graduate School of Medicine.,Department of Clinical Laboratory/Medical Genome Center, National Center for Geriatrics and Gerontology
| | - Kumiko Tanabe
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine
| | - Shinobu Yamaguchi
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine
| | - Tomoyuki Hioki
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine.,Department of Dermatology, Kizawa Memorial Hospital
| | - Junko Tachi
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine.,Department of Pharmacology, Gifu University Graduate School of Medicine
| | | | - Osamu Kozawa
- Department of Pharmacology, Gifu University Graduate School of Medicine
| | - Hiroki Iida
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine
| |
Collapse
|