1
|
Wang L, Chen JH, Zhang YJ, Zhang MB, Zeng T. PPARβ/δ agonist GW0742 mitigates acute liver damage induced by acetaminophen overdose in mice. Toxicol Appl Pharmacol 2025; 494:117180. [PMID: 39617257 DOI: 10.1016/j.taap.2024.117180] [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: 08/05/2024] [Revised: 11/10/2024] [Accepted: 11/26/2024] [Indexed: 12/15/2024]
Abstract
Liver damage caused by acetaminophen (APAP) overdose remains a worldwide medical problem. New therapeutic medicines for APAP poisoning are needed as the efficacy of the only antidote, N-acetyl-cysteine (NAC), significantly decreases if administered after 8 h of APAP intake and massive APAP overdose remains to induce hepatotoxicity despite the timely administration of NAC. Peroxisome proliferator-activated receptor β/δ (PPARβ/δ) possesses versatile roles including regulation of lipid homeostasis and anti-inflammation in the liver. This study aimed to investigate the effects of GW0742, one specific PPARβ/δ agonist, on APAP-caused liver damage in mice. We found that GW0742 (40 mg/kg, i.p.) pretreatment completely blocked the increase of serum aminotransferase activities, hepatocyte necrosis, oxidative stress, and liver inflammation in mice exposed to 300 mg/kg APAP (i.p.). Mechanistically, GW0742 pretreatment significantly suppressed the M1 polarization of liver Kupffer cells and activation of NLRP3 inflammasome. Interestingly, GW0742 remained effective when administered 6 h after APAP exposure, although its efficacy was less pronounced than that administered 6 h before the APAP challenge. Notably, GW0742 exhibited a more profound effect than NAC evidenced by the lower serum alanine transaminase (ALT) level and the improved histopathological manifestation. Furthermore, exposure to APAP for 6 h had resulted in dramatic liver inflammation, while pretreatment with GW0742 prior to APAP exposure did not influence the increase in serum aminotransferase activity and oxidative stress at 2 h after APAP exposure. These results highlight that PPARβ/δ may be a promising therapeutic target for treating APAP-caused acute liver damage probably acting on liver macrophages.
Collapse
Affiliation(s)
- Lin Wang
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Jing-Hui Chen
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yan-Jing Zhang
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Ming-Bao Zhang
- Department of Gastroenterology, The Second Hospital of Shandong University, Jinan, Shandong 250033, China.
| | - Tao Zeng
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
2
|
Jaeschke H, Ramachandran A. Ferroptosis and Intrinsic Drug-induced Liver Injury by Acetaminophen and Other Drugs: A Critical Evaluation and Historical Perspective. J Clin Transl Hepatol 2024; 12:1057-1066. [PMID: 39649034 PMCID: PMC11622198 DOI: 10.14218/jcth.2024.00324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 12/10/2024] Open
Abstract
Drug-induced hepatotoxicity is a significant clinical issue worldwide. Given the limited treatment options for these liver injuries, understanding the mechanisms and modes of cell death is crucial for identifying novel therapeutic targets. For the past 60 years, reactive oxygen species and iron-dependent lipid peroxidation (LPO) have been hypothesized to be involved in many models of acute drug-induced liver injury. However, this mechanism of toxicity was largely abandoned when apoptosis became the primary focus of cell death research. More recently, ferroptosis-a novel, non-apoptotic form of cell death-was identified in NRAS-mutant HT-1080 fibrosarcoma cells exposed to erastin and other NRLs. Ferroptosis is characterized by glutathione depletion and the impairment of glutathione peroxidase 4 activity, which hinders the detoxification of lipid hydroperoxides. These hydroperoxides then serve as substrates for iron-dependent LPO propagation. This cell death mechanism is now receiving widespread attention, extending well beyond its original identification in cancer research, including in the field of drug-induced liver injury. However, concerns arise when such mechanisms are applied across different cell types and disease states without sufficient validation. This review critically evaluated the historical evidence for iron-dependent LPO as a mechanism of drug-induced hepatotoxicity and explored how these earlier findings have led to the current concept of ferroptosis. Overall, the published data support the idea that multi-layered endogenous antioxidant defense mechanisms in the liver limit the occurrence of pathophysiologically relevant LPO under normal conditions. Only when these defense mechanisms are severely compromised does ferroptosis become a significant mode of drug-induced cell death.
Collapse
Affiliation(s)
- Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
3
|
Mischke J, Klein S, Cornberg M, Kraft ARM. MitoTempo treatment as an approach to cure persistent viral infections? Virology 2024; 600:110280. [PMID: 39492087 DOI: 10.1016/j.virol.2024.110280] [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: 06/25/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024]
Abstract
Chronic viral infections are characterized by exhausted virus-specific T cells. Exhaustion is associated with mitochondrial dysfunction, revealing a possible target for treatment. Targeting these metabolic processes may interfere with the exhaustion process of immune cells during infection. It has been shown that the mitochondria-targeted antioxidant MitoTempo could restore hepatitis-B-virus-specific T cells in vitro. Thus, we investigated MitoTempo as a treatment option using the chronic lymphocytic choriomeningitis virus (LCMVcl13) mouse model. MitoTempo treatment of chronically LCMVcl13 infected mice resulted in a transient reduction of LCMV titer. However, no obvious restoration of functional LCMV-specific T cells was observed, beside subtle changes in phenotype of GP33- and NP205-specific T cells. However, these changes did not translate into significantly more functional responses. Our study showed a transient antiviral effect of MitoTempo, but no profound effect on exhausted T cell responses, although further studies are needed to further elucidate the mechanism and use of MitoTempo.
Collapse
Affiliation(s)
- Jasmin Mischke
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; Twincore, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School, Hannover, Germany; German Center for Infection Research (DZIF), Partner-site Hannover-Braunschweig, Hannover, Germany; Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
| | - Sebastian Klein
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; Twincore, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School, Hannover, Germany; German Center for Infection Research (DZIF), Partner-site Hannover-Braunschweig, Hannover, Germany; Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
| | - Markus Cornberg
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; Twincore, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School, Hannover, Germany; German Center for Infection Research (DZIF), Partner-site Hannover-Braunschweig, Hannover, Germany; Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
| | - Anke R M Kraft
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; Twincore, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School, Hannover, Germany; German Center for Infection Research (DZIF), Partner-site Hannover-Braunschweig, Hannover, Germany; Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany.
| |
Collapse
|
4
|
Bannon ST, Decker ST, Erol ME, Fan R, Huang YT, Chung S, Layec G. Mitochondrial free radicals contribute to cigarette smoke condensate-induced impairment of oxidative phosphorylation in the skeletal muscle in situ. Free Radic Biol Med 2024; 224:325-334. [PMID: 39178923 DOI: 10.1016/j.freeradbiomed.2024.08.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/01/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
Oxidative stress plays a critical role in cellular dysfunction associated with cigarette smoke exposure and aging. Some chemicals from tobacco smoke have the potential to amplify mitochondrial ROS (mROS) production, which, in turn, may impair mitochondrial respiratory function. Accordingly, the present study tested the hypothesis that a mitochondria-targeted antioxidant (MitoTEMPO, MT) would attenuate the inhibitory effects of cigarette smoke on skeletal muscle respiratory capacity of middle-aged mice. Specifically, mitochondrial oxidative phosphorylation was assessed using high-resolution respirometry in permeabilized fibers from the fast-twitch gastrocnemius muscle of middle-aged C57Bl/6J mice. Before the assessment of respiration, tissues were incubated for 1hr with a control buffer (CON), cigarette smoke condensate (2 % dilution, SMOKE), or MitoTEMPO (10 μM) combined with cigarette smoke condensate (MT + SMOKE). Cigarette smoke condensate (CSC) decreased maximal-ADP stimulated respiration (CON: 60 ± 15 pmolO2.s-1.mg-1 and SMOKE: 33 ± 8 pmolO2.s-1.mg-1; p = 0.0001), and this effect was attenuated by MT (MT + SMOKE: 41 ± 7 pmolO2.s-1.mg-1; p = 0.02 with SMOKE). Complex-I specific respiration was inhibited by CSC, with no significant effect of MT (p = 0.35). Unlike CON, the addition of glutamate (ΔGlutamate) had an additive effect on respiration in fibers exposed to CSC (CON: 0.9 ± 1.1 pmolO2.s-1.mg-1 and SMOKE: 5.4 ± 3.7 pmolO2.s-1.mg-1; p = 0.008) and MT (MT + SMOKE: 8.2 ± 3.8 pmolO2.s-1.mg-1; p ≤ 0.01). Complex-II specific respiration was inhibited by CSC but was partially restored by MT (p = 0.04 with SMOKE). Maximal uncoupled respiration induced by FCCP was inhibited by CSC, with no significant effect of MT. These findings underscore that mROS contributes to cigarette smoke condensate-induced inhibition of mitochondrial respiration in fast-twitch gastrocnemius muscle fibers of middle-aged mice thus providing a potential target for therapeutic treatment of smoke-related diseases. In addition, this study revealed that CSC largely impaired muscle respiratory capacity by decreasing metabolic flux through mitochondrial pyruvate transporter (MPC) and/or the enzymes upstream of α-ketoglutarate in the Krebs cycle.
Collapse
Affiliation(s)
- Sean T Bannon
- Department of Kinesiology, University of Massachusetts Amherst, MA, USA
| | - Stephen T Decker
- Department of Kinesiology, University of Massachusetts Amherst, MA, USA; Diabetes and Metabolism Research Center, University of Utah, UT, USA
| | - Muhammet Enes Erol
- Department of Kinesiology, University of Massachusetts Amherst, MA, USA; School of Health and Kinesiology, University of Nebraska Omaha, NE, USA
| | - Rong Fan
- Department of Nutrition, University of Massachusetts Amherst, MA, USA
| | - Yu-Ting Huang
- Department of Kinesiology, University of Massachusetts Amherst, MA, USA
| | - Soonkyu Chung
- Department of Nutrition, University of Massachusetts Amherst, MA, USA
| | - Gwenael Layec
- Department of Kinesiology, University of Massachusetts Amherst, MA, USA; School of Health and Kinesiology, University of Nebraska Omaha, NE, USA.
| |
Collapse
|
5
|
Ramachandran A, Akakpo JY, Curry SC, Rumack BH, Jaeschke H. Clinically relevant therapeutic approaches against acetaminophen hepatotoxicity and acute liver failure. Biochem Pharmacol 2024; 228:116056. [PMID: 38346541 PMCID: PMC11315809 DOI: 10.1016/j.bcp.2024.116056] [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: 12/05/2023] [Revised: 01/15/2024] [Accepted: 02/09/2024] [Indexed: 02/17/2024]
Abstract
Liver injury and acute liver failure caused by an acetaminophen (APAP) overdose is a significant clinical problem in western countries. With the introduction of the mouse model of APAP hepatotoxicity in the 1970 s, fundamental mechanisms of cell death were discovered. This included the recognition that part of the APAP dose is metabolized by cytochrome P450 generating a reactive metabolite that is detoxified by glutathione. After the partial depletion of glutathione, the reactive metabolite will covalently bind to sulfhydryl groups of proteins, which is the initiating event of the toxicity. This insight led to the introduction of N-acetyl-L-cysteine, a glutathione precursor, as antidote against APAP overdose in the clinic. Despite substantial progress in our understanding of the pathomechanisms over the last decades viable new antidotes only emerged recently. This review will discuss the background, mechanisms of action, and the clinical prospects of the existing FDA-approved antidote N-acetylcysteine, of several new drug candidates under clinical development [4-methylpyrazole (fomepizole), calmangafodipir] and examples of additional therapeutic targets (Nrf2 activators) and regeneration promoting agents (thrombopoietin mimetics, adenosine A2B receptor agonists, Wharton's Jelly mesenchymal stem cells). Although there are clear limitations of certain therapeutic approaches, there is reason to be optimistic. The substantial progress in the understanding of the pathophysiology of APAP hepatotoxicity led to the consideration of several drugs for development as clinical antidotes against APAP overdose in recent years. Based on the currently available information, it is likely that this will result in additional drugs that could be used as adjunct treatment for N-acetylcysteine.
Collapse
Affiliation(s)
- Anup Ramachandran
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jephte Y Akakpo
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Steven C Curry
- Department of Medical Toxicology, Banner - University Medical Center Phoenix, Phoenix, AZ, USA; Department of Medicine, and Division of Clinical Data Analytics and Decision Support, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA
| | - Barry H Rumack
- Department of Emergency Medicine and Pediatrics, University of Colorado School of Medicine, Denver, CO, USA
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
6
|
Song X, Hao X, Zhu BT. Role of mitochondrial reactive oxygen species in chemically-induced ferroptosis. Free Radic Biol Med 2024; 223:473-492. [PMID: 38992393 DOI: 10.1016/j.freeradbiomed.2024.07.006] [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] [Received: 01/17/2024] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 07/13/2024]
Abstract
Ferroptosis is a form of iron-dependent regulated cell death which is different from apoptosis. Chemically-induced ferroptosis is characterized by an accumulation of lipid reactive oxygen species (ROS) in the cells. A number of earlier studies have suggested the involvement of mitochondrial ROS in ferroptosis, and the present study seeks to further investigate the role of mitochondrial ROS in the induction of chemically-induced ferroptotic cell death. We find that during erastin-induced, glutathione depletion-associated ferroptosis, mitochondrial ROS accumulation is an important late event, which likely is involved in the final execution of ferroptotic cell death. The mitochondrion-originated ROS is found to accumulate in large quantities inside the nuclei during the late phases of erastin-induced ferroptosis. Completion of the late-phase accumulation of mitochondrion-produced ROS inside the nucleus of a cell likely marks an irreversible point in the cell death process. Similarly, accumulation of large amounts of mitochondrion-produced ROS inside the nucleus is also observed in the late phases of RSL3-induced ferroptosis. The results of this study indicate that the mitochondrial ROS play an important role in the final steps of both erastin- and RSL3-induced ferroptotic cell death.
Collapse
Affiliation(s)
- Xiuhan Song
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Xiangyu Hao
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Bao Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China; Shenzhen Bay Laboratory, Shenzhen, 518055, China.
| |
Collapse
|
7
|
Lambrecht R, Jansen J, Rudolf F, El-Mesery M, Caporali S, Amelio I, Stengel F, Brunner T. Drug-induced oxidative stress actively prevents caspase activation and hepatocyte apoptosis. Cell Death Dis 2024; 15:659. [PMID: 39245717 PMCID: PMC11381522 DOI: 10.1038/s41419-024-06998-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/30/2024] [Accepted: 08/13/2024] [Indexed: 09/10/2024]
Abstract
Cell death is a fundamental process in health and disease. Emerging research shows the existence of numerous distinct cell death modalities with similar and intertwined signaling pathways, but resulting in different cellular outcomes, raising the need to understand the decision-making steps during cell death signaling. Paracetamol (Acetaminophen, APAP)-induced hepatocyte death includes several apoptotic processes but eventually is executed by oncotic necrosis without any caspase activation. Here, we studied this paradoxical form of cell death and revealed that APAP not only fails to activate caspases but also strongly impedes their activation upon classical apoptosis induction, thereby shifting apoptosis to necrosis. While APAP intoxication results in massive drop in mitochondrial respiration, low cellular ATP levels could be excluded as an underlying cause of missing apoptosome formation and caspase activation. In contrast, we identified oxidative stress as a key factor in APAP-induced caspase inhibition. Importantly, caspase inhibition and the associated switch from apoptotic to necrotic cell death was reversible through the administration of antioxidants. Thus, exemplified by APAP-induced cell death, our study stresses that cellular redox status is a critical component in the decision-making between apoptotic and necrotic cell death, as it directly affects caspase activity.
Collapse
Affiliation(s)
- Rebekka Lambrecht
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Konstanz, Germany
- Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Jasmin Jansen
- Biochemistry and Mass Spectrometry, Department of Biology, University of Konstanz, Konstanz, Germany
- Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| | - Franziska Rudolf
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Konstanz, Germany
- Collaborative Research Center TRR 353, Konstanz, Germany
| | - Mohamed El-Mesery
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Konstanz, Germany
- Collaborative Research Center TRR 353, Konstanz, Germany
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Sabrina Caporali
- Systems Toxicology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Ivano Amelio
- Collaborative Research Center TRR 353, Konstanz, Germany
- Systems Toxicology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Florian Stengel
- Biochemistry and Mass Spectrometry, Department of Biology, University of Konstanz, Konstanz, Germany
- Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
- Collaborative Research Center TRR 353, Konstanz, Germany
| | - Thomas Brunner
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Konstanz, Germany.
- Collaborative Research Center TRR 353, Konstanz, Germany.
| |
Collapse
|
8
|
Hu J, Nieminen AL, Zhong Z, Lemasters JJ. Role of Mitochondrial Iron Uptake in Acetaminophen Hepatotoxicity. LIVERS 2024; 4:333-351. [PMID: 39554796 PMCID: PMC11567147 DOI: 10.3390/livers4030024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2024] Open
Abstract
Overdose of acetaminophen (APAP) produces fulminant hepatic necrosis. The underlying mechanism of APAP hepatotoxicity involves mitochondrial dysfunction, including mitochondrial oxidant stress and the onset of mitochondrial permeability transition (MPT). Reactive oxygen species (ROS) play an important role in APAP-induced hepatotoxicity, and iron is a critical catalyst for ROS formation. This review summarizes the role of mitochondrial ROS formation in APAP hepatotoxicity and further focuses on the role of iron. Normally, hepatocytes take up Fe3+-transferrin bound to transferrin receptors via endocytosis. Concentrated into lysosomes, the controlled release of iron is required for the mitochondrial biosynthesis of heme and non-heme iron-sulfur clusters. After APAP overdose, the toxic metabolite, NAPQI, damages lysosomes, causing excess iron release and the mitochondrial uptake of Fe2+ by the mitochondrial calcium uniporter (MCU). NAPQI also inhibits mitochondrial respiration to promote ROS formation, including H2O2, with which Fe2+ reacts to form highly reactive •OH through the Fenton reaction. •OH, in turn, causes lipid peroxidation, the formation of toxic aldehydes, induction of the MPT, and ultimately, cell death. Fe2+ also facilitates protein nitration. Targeting pathways of mitochondrial iron movement and consequent iron-dependent mitochondrial ROS formation is a promising strategy to intervene against APAP hepatotoxicity in a clinical setting.
Collapse
Affiliation(s)
- Jiangting Hu
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Anna-Liisa Nieminen
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Zhi Zhong
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - John J Lemasters
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
9
|
Carvalho C, Moreira PI. MitoTempo protects against nε-carboxymethyl lysine-induced mitochondrial dyshomeostasis and neuronal cells injury. Free Radic Biol Med 2024; 220:192-206. [PMID: 38734265 DOI: 10.1016/j.freeradbiomed.2024.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/02/2024] [Accepted: 05/08/2024] [Indexed: 05/13/2024]
Abstract
Enhanced formation of advanced glycation end products (AGEs) is a pivotal factor in diabetes pathophysiology, increasing the risk of diabetic complications. Nε-carboxy-methyl-lysine (CML) is one of the most relevant AGEs found in several tissues including the peripheral blood of diabetic subjects. Despite recognizing diabetes as a risk factor for neurodegenerative diseases and the documented role of mitochondrial abnormalities in this connection, the impact of CML on neuronal mitochondria and its contribution to diabetes-related neurodegeneration remain uncertain. Here, we evaluated the effects of CML in differentiated SH-SY5Y human neuroblastoma cells. Due to the association between mitochondrial dysfunction and increased production of reactive oxygen species (ROS), the possible protective effects of MitoTempo, a mitochondria-targeted antioxidant, were also evaluated. Several parameters were assessed namely cells viability, mitochondrial respiration and membrane potential, ATP and ROS production, Ca2+ levels, mitochondrial biogenesis and dynamics, mito/autophagy, endoplasmic reticulum (ER) stress and amyloidogenic and synaptic integrity markers. CML caused pronounced mitochondrial defects characterized by a significant decrease in mitochondrial respiration, membrane potential, and ATP production and an increase in ROS production. An accumulation of individual mitochondria associated with disrupted mitochondrial networks was also observed. Furthermore, CML caused mitochondrial fusion and a decrease in mitochondrial mass and induced ER stress associated with altered unfolded protein response and Ca2+ dyshomeostasis. Moreover, CML increased the protein levels of β-secretase-1 and amyloid precursor protein, key proteins involved in Alzheimer's Disease pathophysiology. All these effects contributed to the decline in neuronal cells viability. Notable, MitoTempo was able to counteract most of CML-mediated mitochondrial defects and neuronal cells injury and death. Overall, these findings suggest that CML induces pronounced defects in neuronal mitochondria and ER stress, predisposing to neurodegenerative events. More, our observations suggest that MitoTempo holds therapeutic promise in mitigating CML-induced mitochondrial imbalance and neuronal damage and death.
Collapse
Affiliation(s)
- Cristina Carvalho
- Center for Neuroscience and Cell Biology, University of Coimbra (CNC-UC), Portugal; Center for Innovation in Biomedicine and Biotechnology (CIBB), Portugal; Institute for Interdisciplinary Research (III), University of Coimbra, Portugal.
| | - Paula I Moreira
- Center for Neuroscience and Cell Biology, University of Coimbra (CNC-UC), Portugal; Center for Innovation in Biomedicine and Biotechnology (CIBB), Portugal; Institute of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
10
|
Adelusi OB, Etemadi Y, Akakpo JY, Ramachandran A, Jaeschke H. Effect of ferroptosis inhibitors in a murine model of acetaminophen-induced liver injury. J Biochem Mol Toxicol 2024; 38:e23791. [PMID: 39082238 PMCID: PMC11382325 DOI: 10.1002/jbt.23791] [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: 05/11/2024] [Revised: 07/08/2024] [Accepted: 07/19/2024] [Indexed: 08/02/2024]
Abstract
Liver injury caused by acetaminophen (APAP) overdose is the leading cause of acute liver failure in western countries. The mode of APAP-induced cell death has been controversially discussed with ferroptosis emerging as a more recent hypothesis. Ferroptosis is characterized by ferrous iron-catalyzed lipid peroxidation (LPO) causing cell death, which can be prevented by the lipophilic antioxidants ferrostatin-1 and UAMC-3203. To assess the efficacy of these ferroptosis inhibitors, we used two murine models of APAP hepatotoxicity, APAP overdose alone or in combination with FeSO4 in fasted male C57BL/6J mice. APAP triggered severe liver injury in the absence of LPO measured as hepatic malondialdehyde (MDA) levels. In contrast, ferrous iron co-treatment aggravated APAP-induced liver injury and caused extensive LPO. Standard doses of ferrostatin-1 did not affect MDA levels or the injury in both models. In contrast, UAMC-3203 partially protected in both models and reduced LPO in the presence of ferrous iron. However, UAMC-3203 attenuated the translocation of phospho-JNK through downregulation of the mitochondrial anchor protein Sab resulting in reduced mitochondrial dysfunction and liver injury. Thus, APAP toxicity does not involve ferroptosis under normal conditions. The lack of effects of ferroptosis inhibitors in the pathophysiology indicates that ferroptosis signaling pathways are not relevant therapeutic targets.
Collapse
Affiliation(s)
- Olamide B Adelusi
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Yasaman Etemadi
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jephte Y Akakpo
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
11
|
Kar E, Kar F, Can B, Çakır Gündoğdu A, Özbayer C, Koçak FE, Şentürk H. Prophylactic and Therapeutic Efficacy of Boric Acid on Lipopolysaccharide-Induced Liver and Kidney Inflammation in Rats. Biol Trace Elem Res 2024; 202:3701-3713. [PMID: 37910263 DOI: 10.1007/s12011-023-03941-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 10/25/2023] [Indexed: 11/03/2023]
Abstract
In our study, we aimed to examine possible prophylactic (P) or therapeutic (T) effects of boric acid (BA) on lipopolysaccharide (LPS) induced liver and kidney damages. Thirty-two rats were divided into four groups as control, LPS, BAP+LPS, and LPS+BAT. BA was given orally to the rats one hour before the intraperitoneal LPS administration in the BAP+LPS group and one hour after the LPS administration in the LPS+BAT group. Malondialdehyde (MDA), myeloperoxidase (MPO), interleukin-6 (IL-6), IL-10, reduced glutathione (GSH), total oxidant and antioxidant status (TOS and TAS), semaphorin-3A (SEMA3A), cytochrome c (CYCS), and caspase-3 (CASP3) parameters were determined by ELISA method to monitor inflammation, oxidative stress, and apoptosis in the liver and kidney tissues of rats. In addition, alkaline phosphatase (ALP), alanine aminotransferase (ALT), aspartate aminotransferase (AST), urea, creatinine (CREA), C-reactive protein (CRP), gamma glutamyl transferase (GGT), glucose (GLU), sodium (Na), potassium (K), and chlorine (Cl) biochemical parameters were measured in rat serums to monitor liver and kidney functions. Liver and kidney tissues were also examined histopathologically and immunohistochemically. All data were statistically analyzed. Our histological, biochemical, inflammatory, oxidative stress, and apoptotic findings showed that LPS causes serious damage to liver and kidney tissues. Boric acid application brought about significant improvements on the parameters. However, this improvement was seen in the BAP+LPS group, and the results of the LPS+BAT group were insufficient to improve. Our results showed that boric acid administration is effective on severe liver and kidney damage caused by LPS. It has been concluded that prophylactic application is more effective, while therapeutic application is insufficient.
Collapse
Affiliation(s)
- Ezgi Kar
- Training and Research Center, Kutahya Health Sciences University, Kutahya, Turkey.
| | - Fatih Kar
- Department of Medical Biochemistry, Faculty of Medicine, Kutahya Health Sciences University, Kutahya, Turkey
| | - Betül Can
- Department of Medical Biochemistry, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Ayşe Çakır Gündoğdu
- Department of Histology and Embryology, Faculty of Medicine, Kutahya Health Sciences University, Kutahya, Turkey
| | - Cansu Özbayer
- Department of Medical Biology, Faculty of Medicine, Kutahya Health Sciences University, Kutahya, Turkey
| | - Fatma Emel Koçak
- Department of Medical Biochemistry, Faculty of Medicine, Kutahya Health Sciences University, Kutahya, Turkey
| | - Hakan Şentürk
- Department of Biology, Faculty of Art and Sciences, Eskisehir Osmangazi University, Eskisehir, Turkey
| |
Collapse
|
12
|
McGill MR. The Role of Mechanistic Biomarkers in Understanding Acetaminophen Hepatotoxicity in Humans. Drug Metab Dispos 2024; 52:729-739. [PMID: 37918967 PMCID: PMC11257692 DOI: 10.1124/dmd.123.001281] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/18/2023] [Accepted: 10/30/2023] [Indexed: 11/04/2023] Open
Abstract
Our understanding of the fundamental molecular mechanisms of acetaminophen (APAP) hepatotoxicity began in 1973 to 1974, when investigators at the US National Institutes of Health published seminal studies demonstrating conversion of APAP to a reactive metabolite that depletes glutathione and binds to proteins in the liver in mice after overdose. Since then, additional groundbreaking experiments have demonstrated critical roles for mitochondrial damage, oxidative stress, nuclear DNA fragmentation, and necrotic cell death as well. Over the years, some investigators have also attempted to translate these mechanisms to humans using human specimens from APAP overdose patients. This review presents those studies and summarizes what we have learned about APAP hepatotoxicity in humans so far. Overall, the mechanisms of APAP hepatotoxicity in humans strongly resemble those discovered in experimental mouse and cultured hepatocyte models, and emerging biomarkers also suggest similarities in liver repair. The data not only validate the first mechanistic studies of APAP-induced liver injury performed 50 years ago but also demonstrate the human relevance of numerous studies conducted since then. SIGNIFICANCE STATEMENT: Human studies using novel translational, mechanistic biomarkers have confirmed that the fundamental mechanisms of acetaminophen (APAP) hepatotoxicity discovered in rodent models since 1973 are the same in humans. Importantly, these findings have guided the development and understanding of treatments such as N-acetyl-l-cysteine and 4-methylpyrazole over the years. Additional research may improve not only our understanding of APAP overdose pathophysiology in humans but also our ability to predict and treat serious liver injury in patients.
Collapse
Affiliation(s)
- Mitchell R McGill
- Department of Environmental Health Sciences, Fay W. Boozman College of Public Health; Department of Pharmacology and Toxicology, College of Medicine; and Department of Pathology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
13
|
Jaeschke H, Ramachandran A. Central Mechanisms of Acetaminophen Hepatotoxicity: Mitochondrial Dysfunction by Protein Adducts and Oxidant Stress. Drug Metab Dispos 2024; 52:712-721. [PMID: 37567742 PMCID: PMC11257690 DOI: 10.1124/dmd.123.001279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
Acetaminophen (APAP) is an analgesic and antipyretic drug used worldwide, which is safe at therapeutic doses. However, an overdose can induce liver injury and even liver failure. Mechanistic studies in mice beginning with the seminal papers published by B.B. Brodie's group in the 1970s have resulted in important insight into the pathophysiology. Although the metabolic activation of APAP with generation of a reactive metabolite, glutathione depletion, and protein adduct formation are critical initiating events, more recently, mitochondria have come into focus as an important target and decision point of cell death. This review provides a comprehensive overview of the induction of mitochondrial superoxide and peroxynitrite formation and its propagation through a mitogen-activated protein kinase cascade, the mitochondrial permeability transition pore opening caused by iron-catalyzed protein nitration, and the mitochondria-dependent nuclear DNA fragmentation. In addition, the role of adaptive mechanisms that can modulate the pathophysiology, including autophagy, mitophagy, nuclear erythroid 2 p45-related factor 2 activation, and mitochondrial biogenesis, are discussed. Importantly, it is outlined how the mechanisms elucidated in mice translate to human hepatocytes and APAP overdose patients, and how this mechanistic insight explains the mechanism of action of the clinically approved antidote N-acetylcysteine and led to the recent discovery of a novel compound, fomepizole, which is currently under clinical development. SIGNIFICANCE STATEMENT: Acetaminophen (APAP)-induced liver injury is the most frequent cause of acute liver failure in western countries. Extensive mechanistic research over the last several decades has revealed a central role of mitochondria in the pathophysiology of APAP hepatotoxicity. This review article provides a comprehensive discussion of a) mitochondrial protein adducts and oxidative/nitrosative stress, b) mitochondria-regulated nuclear DNA fragmentation, c) adaptive mechanisms to APAP-induced cellular stress, d) translation of cell death mechanisms to overdose patients, and e) mechanism-based antidotes against APAP-induced liver injury.
Collapse
Affiliation(s)
- Hartmut Jaeschke
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
14
|
Blagov AV, Summerhill VI, Sukhorukov VN, Zhigmitova EB, Postnov AY, Orekhov AN. Potential use of antioxidants for the treatment of chronic inflammatory diseases. Front Pharmacol 2024; 15:1378335. [PMID: 38818374 PMCID: PMC11137403 DOI: 10.3389/fphar.2024.1378335] [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: 01/29/2024] [Accepted: 04/26/2024] [Indexed: 06/01/2024] Open
Abstract
The excessive production of various reactive oxidant species over endogenous antioxidant defense mechanisms leads to the development of a state of oxidative stress, with serious biological consequences. The consequences of oxidative stress depend on the balance between the generation of reactive oxidant species and the antioxidant defense and include oxidative damage of biomolecules, disruption of signal transduction, mutation, and cell apoptosis. Accumulating evidence suggests that oxidative stress is involved in the physiopathology of various debilitating illnesses associated with chronic inflammation, including cardiovascular diseases, diabetes, cancer, or neurodegenerative processes, that need continuous pharmacological treatment. Oxidative stress and chronic inflammation are tightly linked pathophysiological processes, one of which can be simply promoted by another. Although, many antioxidant trials have been unsuccessful (some of the trials showed either no effect or even harmful effects) in human patients as a preventive or curative measure, targeting oxidative stress remains an interesting therapeutic approach for the development of new agents to design novel anti-inflammatory drugs with a reliable safety profile. In this regard, several natural antioxidant compounds were explored as potential therapeutic options for the treatment of chronic inflammatory diseases. Several metalloenzymes, such as superoxide dismutase, catalase, and glutathione peroxidase, are among the essential enzymes that maintain the low nanomolar physiological concentrations of superoxide (O2•-) and hydrogen peroxide (H2O2), the major redox signaling molecules, and thus play important roles in the alteration of the redox homeostasis. These enzymes have become a striking source of motivation to design catalytic drugs to enhance the action of these enzymes under pathological conditions related to chronic inflammation. This review is focused on several major representatives of natural and synthetic antioxidants as potential drug candidates for the treatment of chronic inflammatory diseases.
Collapse
Affiliation(s)
| | | | - Vasily N. Sukhorukov
- Institute of General Pathology and Pathophysiology, Moscow, Russia
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Federal State Budgetary Scientific Institution, Petrovsky National Research Centre of Surgery (FSBSI “Petrovsky NRCS”), Moscow, Russia
| | | | - Anton Y. Postnov
- Institute of General Pathology and Pathophysiology, Moscow, Russia
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Federal State Budgetary Scientific Institution, Petrovsky National Research Centre of Surgery (FSBSI “Petrovsky NRCS”), Moscow, Russia
| | - Alexander N. Orekhov
- Institute of General Pathology and Pathophysiology, Moscow, Russia
- Institute for Atherosclerosis Research, Moscow, Russia
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Federal State Budgetary Scientific Institution, Petrovsky National Research Centre of Surgery (FSBSI “Petrovsky NRCS”), Moscow, Russia
| |
Collapse
|
15
|
Elkhawagah AR, Ricci A, Bertero A, Poletto ML, Nervo T, Donato GG, Vincenti L, Martino NA. Supplementation with MitoTEMPO before cryopreservation improves sperm quality and fertility potential of Piedmontese beef bull semen. Front Vet Sci 2024; 11:1376057. [PMID: 38812559 PMCID: PMC11135289 DOI: 10.3389/fvets.2024.1376057] [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: 01/24/2024] [Accepted: 05/03/2024] [Indexed: 05/31/2024] Open
Abstract
The purpose of this study was to improve the quality of frozen-thawed Piedmontese bull semen by incorporating MitoTEMPO (MT) in extended semen before cryopreservation. Semen was collected from 4 fertile bulls, using an artificial vagina, once weekly for 6 consecutive weeks. Semen samples were pooled, diluted with Bullxcell® extender, and supplemented with different concentrations of MT (0 as control, 5, 10, 20, 40, and 80 μM) before cooling, equilibration, and freezing procedures. The frozen-thawed semen was assessed for motility, vitality, acrosome intactness, plasma membrane integrity, DNA integrity, apoptosis, mitochondrial membrane potential, intracellular ROS level and in vitro fertilizing capability. The results showed that MT at concentrations of 10, 20, and 40 μM improved the total, progressive, and rapid motility directly after thawing while, at the highest tested concentration (80 μM), it decreased the progressive and rapid motility after 1, 2, and 3 h of incubation. The sperm kinetics including STR and LIN were noticeably increased at concentrations of 10, 20, and 40 μM directly after thawing (0 h), whereas the MT effect was variable on the other sperm kinetics during the different incubation periods. MitoTEMPO improved the sperm vitality at all tested concentrations, while the acrosomal and DNA integrity were improved at 20 μM and the mitochondrial membrane potentials was increased at 80 μM. The cleavage and blastocyst formation rates were significantly increased by using semen treated with 20 μM MT compared with controls. These findings suggest a potential use of MT mainly at a concentration of 20 μM as an additive in the cryopreservation media of bull semen to improve sperm quality.
Collapse
Affiliation(s)
- Ahmed R. Elkhawagah
- Theriogenology Department, Faculty of Veterinary Medicine, Benha University, Banha, Egypt
| | - Alessandro Ricci
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | - Alessia Bertero
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | | | - Tiziana Nervo
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | - Gian Guido Donato
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | - Leila Vincenti
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | - Nicola Antonio Martino
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
16
|
Haider S, Chakraborty S, Chowdhury G, Chakrabarty A. Opposing Interplay between Nuclear Factor Erythroid 2-Related Factor 2 and Forkhead BoxO 1/3 is Responsible for Sepantronium Bromide's Poor Efficacy and Resistance in Cancer cells: Opportunity for Combination Therapy in Triple Negative Breast Cancer. ACS Pharmacol Transl Sci 2024; 7:1237-1251. [PMID: 38751638 PMCID: PMC11091984 DOI: 10.1021/acsptsci.3c00279] [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: 10/11/2023] [Revised: 03/29/2024] [Accepted: 04/10/2024] [Indexed: 05/18/2024]
Abstract
Survivin, a cancer-cell-specific multifunctional protein, is regulated by many oncogenic signaling pathways and an effective therapeutic target. Although, several types of survivin-targeting agents have been developed over the past few decades, none of them received clinical approval. This could be because survivin expression is tightly controlled by the feedback interaction between different signaling molecules. Of the several signaling pathways that are known to regulate survivin expression, the phosphatidylinositol 3-kinase/AKT serine-threonine kinase/forkhead boxO (PI3K/AKT/FoxO) pathway is well-known for feedback loops constructed by cross-talk among different molecules. Using sepantronium bromide (YM155), the first of its class of survivin-suppressant, we uncovered the existence of an interesting cross-talk between Nuclear Factor Erythroid 2-Related Factor 2 (NRF2) and FoxO transcription factors that also contributes to YM155 resistance in triple negative breast cancer (TNBC) cells. Pharmacological manipulation to interrupt this interaction not only helped restore/enhance the drug-sensitivity but also prompted effective immune clearance of cancer cells. Because the YM155-induced reactive oxygen species (ROS) initiates this feedback, we believe that it will be occurring for many ROS-producing chemotherapeutic agents. Our work provides a rational explanation for the poor efficacy of YM155 compared to standard chemotherapy in clinical trials. Finally, the triple drug combination approach used herein might help reintroducing YM155 into the clinical pipeline, and given the high survivin expression in TNBC cells in general, it could be effective in treating this subtype of breast cancer.
Collapse
Affiliation(s)
- Shaista Haider
- Department
of Life Sciences, Shiv Nadar Institution
of Eminence, Greater Noida Gautam
Buddha Nagar Uttar Pradesh 201314, India
| | - Shayantani Chakraborty
- Department
of Life Sciences, Shiv Nadar Institution
of Eminence, Greater Noida Gautam
Buddha Nagar Uttar Pradesh 201314, India
| | - Goutam Chowdhury
- Independent
Researcher, Greater Noida Gautam Buddha Nagar Uttar Pradesh 201308, India
| | - Anindita Chakrabarty
- Department
of Life Sciences, Shiv Nadar Institution
of Eminence, Greater Noida Gautam
Buddha Nagar Uttar Pradesh 201314, India
| |
Collapse
|
17
|
Zhao L, Jones III JP, Anderson LG, Konsoula Z, Nevison CD, Reissner KJ, Parker W. Acetaminophen causes neurodevelopmental injury in susceptible babies and children: no valid rationale for controversy. Clin Exp Pediatr 2024; 67:126-139. [PMID: 37321575 PMCID: PMC10915458 DOI: 10.3345/cep.2022.01319] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/10/2023] [Accepted: 05/04/2023] [Indexed: 06/17/2023] Open
Abstract
Despite the worldwide acceptance of acetaminophen (APAP) as a necessary medicine in pediatrics, evidence that early exposure to APAP causes neurodevelopmental injury in susceptible babies and children has been mounting for over a decade. The evidence is diverse and includes extensive work with laboratory animals, otherwise unexplained associations, factors associated with APAP metabolism, and limited studies in humans. Although the evidence has reached an overwhelming level and was recently reviewed in detail, controversy persists. This narrative review evaluates some of that controversy. Evidence from the pre- and postpartum periods was considered to avoid controversy raised by consideration of only limited evidence of risks during the prepartum period. Among other issues, the association between APAP use and the prevalence of neurodevelopmental disorders was considered. A systematic review revealed that the use of APAP in the pediatric population was never tracked carefully; however, historical events that affected its use were documented and are sufficient to establish apparent correlations with changes in the prevalence of neurodevelopmental disorders. Moreover, problems with the exclusive reliance on results of meta-analyses of large datasets with limited time frames of drug exposure were reviewed. Furthermore, the evidence of why some children are susceptible to APAPinduced neurodevelopmental injuries was examined. We concluded that available evidence demonstrates that early exposure to APAP causes neurodevelopmental injury in susceptible babies and small children.
Collapse
Affiliation(s)
| | | | | | | | - Cynthia D. Nevison
- Institute for Arctic and Alpine Research, University of Colorado Boulder, Boulder, CO, USA
| | - Kathryn J. Reissner
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, NC, USA
| | | |
Collapse
|
18
|
Lian X, Wang X, Xie Y, Sheng H, He J, Peng T, Xie N, Wang C, Lian Y. ATF5-regulated Mitochondrial Unfolded Protein Response Attenuates Neuronal Damage in Epileptic Rat by Reducing Endoplasmic Reticulum Stress Through Mitochondrial ROS. Neurochem Res 2024; 49:388-401. [PMID: 37847329 DOI: 10.1007/s11064-023-04042-3] [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: 08/03/2023] [Revised: 09/17/2023] [Accepted: 09/28/2023] [Indexed: 10/18/2023]
Abstract
Endoplasmic reticulum (ER) dysfunction caused by excessive ER stress is a crucial mechanism underlying seizures-induced neuronal injury. Studies have shown that mitochondrial reactive oxygen species (ROS) are closely related to ER stress, and our previous study showed that activating transcription factor 5 (ATF5)-regulated mitochondrial unfolded protein response (mtUPR) modulated mitochondrial ROS generation in a hippocampal neuronal culture model of seizures. However, the effects of ATF5-regulated mtUPR on ER stress and the underlying mechanisms remain uncertain in epilepsy. In this study, ATF5 upregulation by lentivirus infection attenuated seizures-induced neuronal damage and apoptosis in a rat model of pilocarpine-induced epilepsy, whereas ATF5 downregulation by lentivirus infection had the opposite effects. ATF5 upregulation potentiated mtUPR by increasing the expression of mitochondrial chaperone heat shock protein 60 (HSP60) and caseinolytic protease proteolytic subunit (ClpP) and reducing mitochondrial ROS generation in pilocarpine-induced seizures in rats. Additionally, upregulation of ATF5 reduced the expression of glucose-regulated protein 78 (GRP78), protein kinase RNA-like endoplasmic reticulum kinase (PERK), activating transcription factor 4 (ATF4), and C/EBP homologous protein (CHOP), suggesting suppression of ER stress; Moreover, ATF5 upregulation attenuated apoptosis-related proteins such as B-cell lymphoma-2 (BCL2) downregulation, BCL2-associated X (BAX) and cleaved-caspase-3 upregulation. However, ATF5 downregulation exerted the opposite effects. Furthermore, pretreatment with the mitochondria-targeted antioxidant mito-TEMPO attenuated the harmful effects of ATF5 downregulation on ER stress and neuronal apoptosis by reducing mitochondrial ROS generation. Overall, our study suggested that ATF5-regulated mtUPR exerted neuroprotective effects against pilocarpine-induced seizures in rats and the underlying mechanisms might involve mitochondrial ROS-mediated ER stress.
Collapse
Affiliation(s)
- Xiaolei Lian
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, Henan, China
- The Academy of Medical Sciences of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaoyi Wang
- Institutes of Biological and Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, Jiangsu, China
| | - Yinyin Xie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, Henan, China
| | - Hanqing Sheng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, Henan, China
| | - Jiao He
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, Henan, China
| | - Tingting Peng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, Henan, China
| | - Nanchang Xie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, Henan, China
| | - Cui Wang
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yajun Lian
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
19
|
Jaeschke H, Ramachandran A. Acetaminophen Hepatotoxicity: Paradigm for Understanding Mechanisms of Drug-Induced Liver Injury. ANNUAL REVIEW OF PATHOLOGY 2024; 19:453-478. [PMID: 38265880 PMCID: PMC11131139 DOI: 10.1146/annurev-pathmechdis-051122-094016] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
Acetaminophen (APAP) overdose is the clinically most relevant drug hepatotoxicity in western countries, and, because of translational relevance of animal models, APAP is mechanistically the most studied drug. This review covers intracellular signaling events starting with drug metabolism and the central role of mitochondrial dysfunction involving oxidant stress and peroxynitrite. Mitochondria-derived endonucleases trigger nuclear DNA fragmentation, the point of no return for cell death. In addition, adaptive mechanisms that limit cell death are discussed including autophagy, mitochondrial morphology changes, and biogenesis. Extensive evidence supports oncotic necrosis as the mode of cell death; however, a partial overlap with signaling events of apoptosis, ferroptosis, and pyroptosis is the basis for controversial discussions. Furthermore, an update on sterile inflammation in injury and repair with activation of Kupffer cells, monocyte-derived macrophages, and neutrophils is provided. Understanding these mechanisms of cell death led to discovery of N-acetylcysteine and recently fomepizole as effective antidotes against APAP toxicity.
Collapse
Affiliation(s)
- Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA; ,
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA; ,
| |
Collapse
|
20
|
Qi XY, Yuan JD, Liu ZY, Jiang XQ, Zhang Q, Zhang SL, Zhao L, Ke LY, Zhang CY, Li Y, Zhang LY, Xu QQ, Liu ZH, Sun JT, Jin JX. Sirtuin 3-mediated deacetylation of superoxide dismutase 2 ameliorates sodium fluoride-induced mitochondrial dysfunction in porcine oocytes. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 908:168306. [PMID: 37944611 DOI: 10.1016/j.scitotenv.2023.168306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023]
Abstract
Fluoride exerts detrimental effects on germ cells and increases the infertility rate in women. Nevertheless, the precise mechanisms behind the developmental abnormalities caused by fluoride in oocytes remain poorly comprehended. The current study, we established mitochondrial damage model in oocytes via 50 μg/mL sodium fluoride (NaF) supplementation. We then examined the effects of honokiol in preventing mitochondrial deficits caused by NaF and investigated the mechanisms through which honokiol protects oocytes. The findings investigated that NaF increased levels of mitochondrial reactive oxygen species (mtROS) and hindered mitochondrial function, as evidenced by the dissipation of mitochondrial membrane potential, abnormal expression of mitochondrial DNA copy numbers, and mtDNA harm in oocytes. mtROS scavenging using Mito-TEMPO alleviated oxidative damage in mitochondria and restored the oocyte developmental competence. Superoxide dismutase 2 (SOD2) acetylation was significantly increased, whereas sirtuin 3 (SIRT3) expression was decreased in NaF-treated oocytes. The addition of honokiol helped in the deacetylation of SOD2 at K122 through SIRT3, resulting in the removal of excessive mtROS and the recovery of mitochondrial function. Therefore, SIRT3/SOD2 pathway aids honokiol in mitigating fluoride-induced mitochondrial dysfunction. Overall, honokiol improved the mitochondrial harm caused by NaF by controlling mtROS and mitochondrial function, with the SIRT3/SOD2 pathway having an important function. These findings suggest honokiol as a potential therapeutic strategy for NaF-induced oocyte development and mitochondrial deficits.
Collapse
Affiliation(s)
- Xin-Yue Qi
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Jin-Dong Yuan
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Zi-Yu Liu
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Xi-Qing Jiang
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Qi Zhang
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Shan-Long Zhang
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Lu Zhao
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Ling-Yan Ke
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Chen-Yuan Zhang
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Yan Li
- School of Environmental Science and Engineering, Yancheng Institute of Technology, Yancheng, China
| | - Lu-Yan Zhang
- School of Environmental Science and Engineering, Yancheng Institute of Technology, Yancheng, China
| | - Qian-Qian Xu
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Zhong-Hua Liu
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, China.
| | - Jing-Tao Sun
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, China.
| | - Jun-Xue Jin
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, China.
| |
Collapse
|
21
|
Lambrecht R, Rudolf F, Ückert AK, Sladky VC, Phan TS, Jansen J, Naim S, Kaufmann T, Keogh A, Kirschnek S, Mangerich A, Stengel F, Leist M, Villunger A, Brunner T. Non-canonical BIM-regulated energy metabolism determines drug-induced liver necrosis. Cell Death Differ 2024; 31:119-131. [PMID: 38001256 PMCID: PMC10781779 DOI: 10.1038/s41418-023-01245-7] [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: 02/16/2023] [Revised: 11/02/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Paracetamol (acetaminophen, APAP) overdose severely damages mitochondria and triggers several apoptotic processes in hepatocytes, but the final outcome is fulminant necrotic cell death, resulting in acute liver failure and mortality. Here, we studied this switch of cell death modes and demonstrate a non-canonical role of the apoptosis-regulating BCL-2 homolog BIM/Bcl2l11 in promoting necrosis by regulating cellular bioenergetics. BIM deficiency enhanced total ATP production and shifted the bioenergetic profile towards glycolysis, resulting in persistent protection from APAP-induced liver injury. Modulation of glucose levels and deletion of Mitofusins confirmed that severe APAP toxicity occurs only in cells dependent on oxidative phosphorylation. Glycolytic hepatocytes maintained elevated ATP levels and reduced ROS, which enabled lysosomal recycling of damaged mitochondria by mitophagy. The present study highlights how metabolism and bioenergetics affect drug-induced liver toxicity, and identifies BIM as important regulator of glycolysis, mitochondrial respiration, and oxidative stress signaling.
Collapse
Affiliation(s)
- Rebekka Lambrecht
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78464, Konstanz, Germany
| | - Franziska Rudolf
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78464, Konstanz, Germany
| | - Anna-Katharina Ückert
- In vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78464, Konstanz, Germany
| | - Valentina C Sladky
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria
| | - Truong San Phan
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78464, Konstanz, Germany
| | - Jasmin Jansen
- Biochemistry and Mass Spectrometry, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78464, Konstanz, Germany
| | - Samara Naim
- Institute of Pharmacology, University of Bern, Inselspital, Bern University Hospital, INO-F, Freiburgstrasse 16C, 3010, Bern, Switzerland
| | - Thomas Kaufmann
- Institute of Pharmacology, University of Bern, Inselspital, Bern University Hospital, INO-F, Freiburgstrasse 16C, 3010, Bern, Switzerland
| | - Adrian Keogh
- Visceral and Transplantation Surgery, Department of Clinical Research, Inselspital, Bern University Hospital, 3008, Bern, Switzerland
| | - Susanne Kirschnek
- Faculty of Medicine, Institute of Medical Microbiology and Hygiene, Medical Center, University of Freiburg, 79104, Freiburg, Germany
| | - Aswin Mangerich
- Nutritional Toxicology, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
| | - Florian Stengel
- Biochemistry and Mass Spectrometry, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78464, Konstanz, Germany
| | - Marcel Leist
- In vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78464, Konstanz, Germany
| | - Andreas Villunger
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria
- The Research Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Lazarettgasse 14, 1090, Vienna, Austria
- Ludwig Boltzman Institute for Rare and Undiagnosed Diseases (LBI-RUD), Lazarettgasse 14, 1090, Vienna, Austria
| | - Thomas Brunner
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78464, Konstanz, Germany.
| |
Collapse
|
22
|
Tüylü Küçükkılınç T, Ercan A. Phenelzine protects against acetaminophen induced apoptosis in HepG2 cells. Drug Chem Toxicol 2024; 47:81-89. [PMID: 37246945 DOI: 10.1080/01480545.2023.2217696] [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: 12/22/2022] [Revised: 04/04/2023] [Accepted: 04/08/2023] [Indexed: 05/30/2023]
Abstract
Acetaminophen (APAP) overdosing is the most common cause of drug-induced liver failure. Despite extensive study, N-acetylcysteine is currently the only antidote utilized for treatment. The purpose of this study was to evaluate the effect and mechanisms of phenelzine, an FDA-approved antidepressant, on APAP-induced toxicity in HepG2 cells. The human liver hepatocellular cell line HepG2 was used to investigate APAP-induced cytotoxicity. The protective effects of phenelzine were determined by examining the cell viability, combination index calculation, Caspase 3/7 activation, Cytochrome c release, H2O2 levels, NO levels, GSH activity, PERK protein levels, and pathway enrichment analysis. Elevated H2O2 production and decreased glutathione (GSH) levels were indicators of APAP-induced oxidative stress. The combination index of 2.04 indicated that phenelzine had an antagonistic effect on APAP-induced toxicity. When compared to APAP alone, phenelzine treatment considerably reduced caspase 3/7 activation, cytochrome c release, and H2O2 generation. However, phenelzine had minimal effect on NO and GSH levels and did not alleviate ER stress. Pathway enrichment analysis revealed a potential connection between APAP toxicity and phenelzine metabolism. These findings suggested that phenelzine's protective effect against APAP-induced cytotoxicity could be attributed to the drug's capacity to reduce APAP-mediated apoptotic signaling.
Collapse
Affiliation(s)
| | - Ayşe Ercan
- Department of Biochemistry, Hacettepe University Faculty of Pharmacy, Ankara, Turkey
| |
Collapse
|
23
|
Parker W, Anderson LG, Jones JP, Anderson R, Williamson L, Bono-Lunn D, Konsoula Z. The Dangers of Acetaminophen for Neurodevelopment Outweigh Scant Evidence for Long-Term Benefits. CHILDREN (BASEL, SWITZERLAND) 2023; 11:44. [PMID: 38255358 PMCID: PMC10814214 DOI: 10.3390/children11010044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/20/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024]
Abstract
Based on available data that include approximately 20 lines of evidence from studies in laboratory animal models, observations in humans, correlations in time, and pharmacological/toxicological considerations, it has been concluded without reasonable doubt and with no evidence to the contrary that exposure of susceptible babies and children to acetaminophen (paracetamol) induces many, if not most, cases of autism spectrum disorder (ASD). However, the relative number of cases of ASD that might be induced by acetaminophen has not yet been estimated. Here, we examine a variety of evidence, including the acetaminophen-induced reduction of social awareness in adults, the prevalence of ASD through time, and crude estimates of the relative number of ASD cases induced by acetaminophen during various periods of neurodevelopment. We conclude that the very early postpartum period poses the greatest risk for acetaminophen-induced ASD, and that nearly ubiquitous use of acetaminophen during early development could conceivably be responsible for the induction in the vast majority, perhaps 90% or more, of all cases of ASD. Despite over a decade of accumulating evidence that acetaminophen is harmful for neurodevelopment, numerous studies demonstrate that acetaminophen is frequently administered to children in excess of currently approved amounts and under conditions in which it provides no benefit. Further, studies have failed to demonstrate long-term benefits of acetaminophen for the pediatric population, leaving no valid rationale for continued use of the drug in that population given its risks to neurodevelopment.
Collapse
Affiliation(s)
- William Parker
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, NC 27599, USA
- WPLab, Inc., Durham, NC 27707, USA
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | - Lauren Williamson
- Department of Biological Sciences, Northern Kentucky University, Highland Heights, KY 41099, USA;
| | - Dillan Bono-Lunn
- Department of Public Policy, University of North Carolina, Chapel Hill, NC 27599, USA;
| | | |
Collapse
|
24
|
Peng C, Luo J, Wang K, Li J, Ma Y, Li J, Yang H, Chen T, Zhang G, Ji X, Liao Y, Lin H, Ji Z. Iridium metal complex targeting oxidation resistance 1 protein attenuates spinal cord injury by inhibiting oxidative stress-associated reactive oxygen species. Redox Biol 2023; 67:102913. [PMID: 37857001 PMCID: PMC10587759 DOI: 10.1016/j.redox.2023.102913] [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: 07/10/2023] [Revised: 09/29/2023] [Accepted: 10/01/2023] [Indexed: 10/21/2023] Open
Abstract
Oxidative stress is a key factor leading to profound neurological deficits following spinal cord injury (SCI). In this study, we present the development and potential application of an iridium (iii) complex, (CpxbiPh) Ir (N^N) Cl, where CpxbiPh represents 1-biphenyl-2,3,4,5-tetramethyl cyclopentadienyl, and N^N denotes 2-(3-(4-nitrophenyl)-1H-1,2,4-triazol-5-yl) pyridine chelating agents, to address this challenge through a mechanism governed by the regulation of an antioxidant protein. This iridium complex, IrPHtz, can modulate the Oxidation Resistance 1 (OXR1) protein levels within spinal cord tissues, thus showcasing its antioxidative potential. By eliminating reactive oxygen species (ROS) and preventing apoptosis, the IrPHtz demonstrated neuroprotective and neural healing characteristics on injured neurons. Our molecular docking analysis unveiled the presence of π stacking within the IrPHtz-OXR1 complex, an interaction that enhanced OXR1 expression, subsequently diminishing oxidative stress, thwarting neuroinflammation, and averting neuronal apoptosis. Furthermore, in in vivo experimentation with SCI-afflicted mice, IrPHtz was efficacious in shielding spinal cord neurons, promoting their regrowth, restoring electrical signaling, and improving motor performance. Collectively, these findings underscore the potential of employing the iridium metal complex in a novel, protein-regulated antioxidant strategy, presenting a promising avenue for therapeutic intervention in SCI.
Collapse
Affiliation(s)
- Cheng Peng
- Department of Orthopedics, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Jianxian Luo
- Department of Orthopedics, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Ke Wang
- Department of Orthopedics, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Jianping Li
- Department of Anatomy, Shaoyang University Puai Medical College, Shaoyang, Hunan, 422099, China
| | - Yanming Ma
- Department of Orthopedics, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Juanjuan Li
- Guangdong Key Laboratory of Urology and Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Hua Yang
- Department of Orthopedics, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Tianjun Chen
- Department of Orthopedics, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Guowei Zhang
- Department of Orthopedics, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Xin Ji
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China.
| | - Yuhui Liao
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital, Southern Medical University, Guangzhou, 510091, China.
| | - Hongsheng Lin
- Department of Orthopedics, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, 510632, China.
| | - Zhisheng Ji
- Department of Orthopedics, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, 510632, China.
| |
Collapse
|
25
|
Yang H, Chen J, Li J. Isolation, culture, and delivery considerations for the use of mesenchymal stem cells in potential therapies for acute liver failure. Front Immunol 2023; 14:1243220. [PMID: 37744328 PMCID: PMC10513107 DOI: 10.3389/fimmu.2023.1243220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/18/2023] [Indexed: 09/26/2023] Open
Abstract
Acute liver failure (ALF) is a high-mortality syndrome for which liver transplantation is considered the only effective treatment option. A shortage of donor organs, high costs and surgical complications associated with immune rejection constrain the therapeutic effects of liver transplantation. Recently, mesenchymal stem cell (MSC) therapy was recognized as an alternative strategy for liver transplantation. Bone marrow mesenchymal stem cells (BMSCs) have been used in clinical trials of several liver diseases due to their ease of acquisition, strong proliferation ability, multipotent differentiation, homing to the lesion site, low immunogenicity and anti-inflammatory and antifibrotic effects. In this review, we comprehensively summarized the harvest and culture expansion strategies for BMSCs, the development of animal models of ALF of different aetiologies, the critical mechanisms of BMSC therapy for ALF and the challenge of clinical application.
Collapse
Affiliation(s)
| | | | - Jun 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, Hangzhou, China
| |
Collapse
|
26
|
Ali Hassan H, Banchi P, Domain G, Vanderheyden L, Prochowska S, Nizański W, Van Soom A. Mito-Tempo improves acrosome integrity of frozen-thawed epididymal spermatozoa in tomcats. Front Vet Sci 2023; 10:1170347. [PMID: 37609058 PMCID: PMC10440435 DOI: 10.3389/fvets.2023.1170347] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 06/01/2023] [Indexed: 08/24/2023] Open
Abstract
Introduction In tomcats, epididymal spermatozoa provide an additional source of male gametes available for cryopreservation. While this procedure is feasible, the survival rate and motility of epididymal cat spermatozoa are both low after thawing. Cryopreservation is known to induce oxidative stress in spermatozoa, with mitochondria and the plasma membrane being the two major generation sites, and an imbalanced presence of free radicals is a possible cause for this low survival rate. Different antioxidants have been tested before for their effect on cryopreserved cat spermatozoa quality, with varying results. Here, we used Mito-Tempo, which is a synthetic mitochondria-targeted antioxidant and a specific scavenger of the mitochondrial superoxide system. By supplementing Mito-Tempo with the freezing extender, we aimed to improve the sperm quality of frozen-thawed cat epididymal spermatozoa. Methods Epididymal spermatozoa obtained from twelve tomcats were assessed for motility and concentration. Prior to freezing, samples were diluted in TRIS buffered extender with egg yolk and glycerol and divided into five aliquots supplemented with 0 (control), 0.5, 5, 50, and 1005M of Mito-Tempo. After thawing, sperm motility, concentration, morphology, plasma membrane integrity, acrosome integrity, and mitochondrial membrane potential were evaluated. A Friedman rank sum test with a Bonferroni post-hoc test was used to determine statistical in-between group differences in post-thaw semen parameters. Results and discussion The results indicated a slight improvement in acrosome integrity across all groups that were supplemented with Mito-Tempo, with the group that received 55M of Mito-Tempo showing the greatest improvement [(median of 67.99%, IQR of 5.55) compared to the control group (median of 65.33%, IQR of 7.75; P = 0.05)]. For all other sperm parameters, no significant differences (P > 0.05) were detected between different Mito-Tempo concentrations. These findings highlight the protective effect of Mito-Tempo on acrosome integrity and suggest that 55M is the most effective concentration for maintaining acrosome integrity. Since Mito-Tempo has shown a positive effect on multiple sperm parameters in other species, such as men, boars, roosters, rams, and bulls, we need to conclude that species-specificity may play a role here.
Collapse
Affiliation(s)
- Hiba Ali Hassan
- Reproductive Biology Unit, Faculty of Veterinary Medicine, Department of Internal Medicine, Reproduction and Population Medicine, Ghent University, Ghent, Merelbeke, Belgium
| | - Penelope Banchi
- Reproductive Biology Unit, Faculty of Veterinary Medicine, Department of Internal Medicine, Reproduction and Population Medicine, Ghent University, Ghent, Merelbeke, Belgium
- Department of Veterinary Sciences, Faculty of Veterinary Medicine, University of Turin, Grugliasco, Italy
| | - Guillaume Domain
- Reproductive Biology Unit, Faculty of Veterinary Medicine, Department of Internal Medicine, Reproduction and Population Medicine, Ghent University, Ghent, Merelbeke, Belgium
| | - Leen Vanderheyden
- Reproductive Biology Unit, Faculty of Veterinary Medicine, Department of Internal Medicine, Reproduction and Population Medicine, Ghent University, Ghent, Merelbeke, Belgium
| | - Sylwia Prochowska
- Department of Reproduction and Clinic of Farm Animals, Faculty of Veterinary Medicine, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Wojciech Nizański
- Department of Reproduction and Clinic of Farm Animals, Faculty of Veterinary Medicine, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Ann Van Soom
- Reproductive Biology Unit, Faculty of Veterinary Medicine, Department of Internal Medicine, Reproduction and Population Medicine, Ghent University, Ghent, Merelbeke, Belgium
| |
Collapse
|
27
|
Abstract
Mitochondria are critical organelles responsible for the maintenance of cellular energy homeostasis. Thus, their dysfunction can have severe consequences in cells responsible for energy-intensive metabolic function, such as hepatocytes. Extensive research over the last decades have identified compromised mitochondrial function as a central feature in the pathophysiology of liver injury induced by an acetaminophen (APAP) overdose, the most common cause of acute liver failure in the United States. While hepatocyte mitochondrial oxidative and nitrosative stress coupled with induction of the mitochondrial permeability transition are well recognized after an APAP overdose, recent studies have revealed additional details about the organelle's role in APAP pathophysiology. This concise review highlights these new advances, which establish the central role of the mitochondria in APAP pathophysiology, and places them in the context of earlier information in the literature. Adaptive alterations in mitochondrial morphology as well as the role of cellular iron in mitochondrial dysfunction and the organelle's importance in liver recovery after APAP-induced injury will be discussed.
Collapse
|
28
|
Lee J, Ha J, Kim JH, Seo D, Kim M, Lee Y, Park SS, Choi D, Park JS, Lee YJ, Yang S, Yang KM, Jung SM, Hong S, Koo SH, Bae YS, Kim SJ, Park SH. Peli3 ablation ameliorates acetaminophen-induced liver injury through inhibition of GSK3β phosphorylation and mitochondrial translocation. Exp Mol Med 2023; 55:1218-1231. [PMID: 37258579 PMCID: PMC10318043 DOI: 10.1038/s12276-023-01009-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 02/07/2023] [Accepted: 03/15/2023] [Indexed: 06/02/2023] Open
Abstract
The signaling pathways governing acetaminophen (APAP)-induced liver injury have been extensively studied. However, little is known about the ubiquitin-modifying enzymes needed for the regulation of APAP-induced liver injury. Here, we examined whether the Pellino3 protein, which has E3 ligase activity, is needed for APAP-induced liver injury and subsequently explored its molecular mechanism. Whole-body Peli3-/- knockout (KO) and adenovirus-mediated Peli3 knockdown (KD) mice showed reduced levels of centrilobular cell death, infiltration of immune cells, and biomarkers of liver injury, such as alanine aminotransferase (ALT) and aspartate aminotransferase (AST), upon APAP treatment compared to wild-type (WT) mice. Peli3 deficiency in primary hepatocytes decreased mitochondrial and lysosomal damage and reduced the mitochondrial reactive oxygen species (ROS) levels. In addition, the levels of phosphorylation at serine 9 in the cytoplasm and mitochondrial translocation of GSK3β were decreased in primary hepatocytes obtained from Peli3-/- KO mice, and these reductions were accompanied by decreases in JNK phosphorylation and mitochondrial translocation. Pellino3 bound more strongly to GSK3β compared with JNK1 and JNK2 and induced the lysine 63 (K63)-mediated polyubiquitination of GSK3β. In rescue experiments, the ectopic expression of wild-type Pellino3 in Peli3-/- KO hepatocytes restored the mitochondrial translocation of GSK3β, but this restoration was not obtained with expression of a catalytically inactive mutant of Pellino3. These findings are the first to suggest a mechanistic link between Pellino3 and APAP-induced liver injury through the modulation of GSK3β polyubiquitination.
Collapse
Affiliation(s)
- Jaewon Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jihoon Ha
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jun-Hyeong Kim
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- KoBio Labs, Seongnam, 13488, Republic of Korea
| | - Dongyeob Seo
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Minbeom Kim
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Yerin Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Seong Shil Park
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Dahee Choi
- Department of Life Science, Korea University, Seoul, 02841, Republic of Korea
| | - Jin Seok Park
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Young Jae Lee
- Department of Biochemistry, Gachon University School of Medicine, Incheon, 21999, Republic of Korea
| | - Siyoung Yang
- Department of Pharmacology, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
- SRC Center for Immune Research on Non-lymphoid Organs, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | | | - Su Myung Jung
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Suntaek Hong
- Department of Biochemistry, Gachon University School of Medicine, Incheon, 21999, Republic of Korea
| | - Seung-Hoi Koo
- Department of Life Science, Korea University, Seoul, 02841, Republic of Korea
| | - Yong-Soo Bae
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- SRC Center for Immune Research on Non-lymphoid Organs, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Seong-Jin Kim
- Medpacto Inc., Seoul, 06668, Republic of Korea.
- GILO Institute, GILO Foundation, Seoul, 06668, Republic of Korea.
| | - Seok Hee Park
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
- SRC Center for Immune Research on Non-lymphoid Organs, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
29
|
Yu B, Zhang Y, Wang T, Guo J, Kong C, Chen Z, Ma X, Qiu T. MAPK Signaling Pathways in Hepatic Ischemia/Reperfusion Injury. J Inflamm Res 2023; 16:1405-1418. [PMID: 37012971 PMCID: PMC10065871 DOI: 10.2147/jir.s396604] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/16/2023] [Indexed: 03/29/2023] Open
Abstract
The mitogen-activated protein kinase signaling pathway can be activated by a variety of growth factors, cytokines, and hormones, and mediates numerous intracellular signals related to cellular activities, including cell proliferation, motility, and differentiation. It has been widely studied in the occurrence and development of inflammation and tumor. Hepatic ischemia-reperfusion injury (HIRI) is a common pathophysiological phenomenon that occurs in surgical procedures such as lobectomy and liver transplantation, which is characterized by severe inflammatory reaction after ischemia and reperfusion. In this review, we mainly discuss the role of p38, ERK1/2, JNK in MAPK family and TAK1 and ASK1 in MAPKKK family in HIRI, and try to find an effective treatment for HIRI.
Collapse
Affiliation(s)
- Bo Yu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - Yalong Zhang
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - Tianyu Wang
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - Jiayu Guo
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - Chenyang Kong
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - Zhongbao Chen
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - Xiaoxiong Ma
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - Tao Qiu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
- Correspondence: Tao Qiu, Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China, Tel +86-13995632367, Email
| |
Collapse
|
30
|
Lin Z, Wang H, Song J, Xu G, Lu F, Ma X, Xia X, Jiang J, Zou F. The role of mitochondrial fission in intervertebral disc degeneration. Osteoarthritis Cartilage 2023; 31:158-166. [PMID: 36375758 DOI: 10.1016/j.joca.2022.10.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/06/2022] [Accepted: 10/31/2022] [Indexed: 11/13/2022]
Abstract
Low back pain (LBP) is an extremely common disorder and is a major cause of disability globally. Intervertebral disc degeneration (IVDD) is the main contributor to LBP. Nevertheless, the specific mechanisms underlying the pathogenesis of IVDD remain unclear. Mitochondria are highly dynamic organelles that continuously undergo fusion and fission, known as mitochondrial dynamics. Accumulating evidence has revealed that aberrantly activated mitochondrial fission leads to mitochondrial fragmentation and dysfunction, which are involved in the development and progression of IVDD. To date, research into mitochondrial dynamics in IVDD is at an early stage. The present narrative review aims to summarize the most recent findings about the role of mitochondrial fission in the pathogenesis of IVDD.
Collapse
Affiliation(s)
- Z Lin
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - H Wang
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - J Song
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - G Xu
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - F Lu
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - X Ma
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - X Xia
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - J Jiang
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - F Zou
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
31
|
Luo G, Huang L, Zhang Z. The molecular mechanisms of acetaminophen-induced hepatotoxicity and its potential therapeutic targets. Exp Biol Med (Maywood) 2023; 248:412-424. [PMID: 36670547 DOI: 10.1177/15353702221147563] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Acetaminophen (APAP), a widely used antipyretic and analgesic drug in clinics, is relatively safe at therapeutic doses; however, APAP overdose may lead to fatal acute liver injury. Currently, N-acetylcysteine (NAC) is clinically used as the main antidote for APAP poisoning, but its therapeutic effect remains limited owing to rapid disease progression and the general diagnosis of advanced poisoning. As is well known, APAP-induced hepatotoxicity (AIH) is mainly caused by the toxic metabolite N-acetyl-p-benzoquinone imine (NAPQI), and the toxic mechanisms of AIH are complicated. Several cellular processes are involved in the pathogenesis of AIH, including liver metabolism, mitochondrial oxidative stress and dysfunction, sterile inflammation, endoplasmic reticulum stress, autophagy, and microcirculation dysfunction. Mitochondrial oxidative stress and dysfunction are the major cellular events associated with APAP-induced liver injury. Many biomolecules involved in these biological processes are potential therapeutic targets for AIH. Therefore, there is an urgent need to comprehensively clarify the molecular mechanisms underlying AIH and to explore novel therapeutic strategies. This review summarizes the various cellular events involved in AIH and discusses their potential therapeutic targets, with the aim of providing new ideas for the treatment of AIH.
Collapse
Affiliation(s)
- Guangwen Luo
- Jinhua Municipal Central Hospital, Jinhua 321000, China
| | - Lili Huang
- Ningbo Medical Center Lihuili Hospital, Ningbo 315040, China
| | - Zhaowei Zhang
- Jinhua Municipal Central Hospital, Jinhua 321000, China
| |
Collapse
|
32
|
Abstract
Acetaminophen (APAP) is a widely used pain reliever that can cause liver injury or liver failure in response to an overdose. Understanding the mechanisms of APAP-induced cell death is critical for identifying new therapeutic targets. In this respect it was hypothesized that hepatocytes die by oncotic necrosis, apoptosis, necroptosis, ferroptosis and more recently pyroptosis. The latter cell death is characterized by caspase-dependent gasdermin cleavage into a C-terminal and an N-terminal fragment, which forms pores in the plasma membrane. The gasdermin pores can release potassium, interleukin-1β (IL-1β), IL-18, and other small molecules in a sublytic phase, which can be the main function of the pores in certain cell types such as inflammatory cells. Alternatively, the process can progress to full lysis of the cell (pyroptosis) with extensive cell contents release. This review discusses the experimental evidence for the involvement of pyroptosis in APAP hepatotoxicity as well as the arguments against pyroptosis as a relevant mechanism of APAP-induced cell death in hepatocytes. Based on the critical evaluation of the currently available literature and understanding of the pathophysiology, it can be concluded that pyroptotic cell death is unlikely to be a relevant contributor to APAP-induced liver injury.
Collapse
Affiliation(s)
- Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - David S. Umbaugh
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
33
|
Patel E, Jones Iii JP, Bono-Lunn D, Kuchibhatla M, Palkar A, Cendejas Hernandez J, Sarafian JT, Lawton VG, Anderson LG, Konsoula Z, Reissner KJ, Parker W. The safety of pediatric use of paracetamol (acetaminophen): a narrative review of direct and indirect evidence. Minerva Pediatr (Torino) 2022; 74:774-788. [PMID: 35822581 DOI: 10.23736/s2724-5276.22.06932-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Paracetamol (acetaminophen) use during pregnancy and early childhood was accepted as safe in the 1970s, but is now a subject of considerable concern. Careful analysis shows that initial acceptance of the drug was based on the false assumption that drug interactions in babies and adults are the same, and on a complete absence of knowledge regarding the impact of the drug on brain development. At least fourteen epidemiological studies now indicate that prenatal exposure to paracetamol is associated with neurodevelopmental problems. Based on these studies, it can be concluded that prenatal exposure to paracetamol causes statistically significant risks of developmental delays, attention deficit hyperactivity disorder, and a subtype of autism spectrum disorder (ASD) associated with hyperkinetic behavior. In contrast, data regarding postnatal exposure to paracetamol are limited, and several factors impede a classic multivariate analysis of epidemiologic data to resolve the issue. However, circumstantial evidence regarding postnatal exposure to the drug is abundant, and includes at least three otherwise unexplained temporal relationships, data from laboratory animal studies, several miscellaneous and otherwise unexplained correlations, and a lack of alternative suspects that fit the evidence-derived profile. Based on this evidence, it can be concluded without any reasonable doubt that oxidative stress puts some babies and children at risk of paracetamol-induced neurodevelopmental injury, and that postnatal exposure to paracetamol in those susceptible babies and children is responsible for many if not most cases of ASD.
Collapse
Affiliation(s)
| | - John P Jones Iii
- WPLab, Inc. Durham, NC, USA.,Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Dillan Bono-Lunn
- Departments of Public Policy, University of North Carolina, Chapel Hill, NC, USA
| | - Maragatha Kuchibhatla
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, USA
| | - Antara Palkar
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | | | - Joshua T Sarafian
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Victoria G Lawton
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Lauren G Anderson
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | | | - Kathryn J Reissner
- Department of Psychology, University of North Carolina, Chapel Hill, NC, USA.,Department of Neuroscience, University of North Carolina, Chapel Hill, NC, USA
| | - William Parker
- WPLab, Inc. Durham, NC, USA - .,Department of Surgery, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
34
|
Virgana R, Gunadi JW, Atik N, Limdawati K, Jasaputra DK, Wahyudianingsih R, Suardi NNA, Soetadji RS, Goenawan H, Lesmana R, Kartasasmita AS. MitoTEMPOL modulates mitophagy and histopathology of Wistar rat liver after streptozotocin injection. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:1382-1388. [PMID: 36474569 PMCID: PMC9699945 DOI: 10.22038/ijbms.2022.65285.14375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 09/14/2022] [Indexed: 01/25/2023]
Abstract
OBJECTIVES This study aims to explore the effect of mitoTEMPOL on histopathology, lipid droplet, and mitophagy gene expression of Wistar rat's liver after injection of streptozotocin (STZ). MATERIALS AND METHODS Twenty male Wistar rats were divided into 4 groups: Control (n=5); 100 mg/kg BW/day mitoTEMPOL orally (n=5); 50 mg/kg BW STZ intraperitoneal injection (n=5); and mitoTEMPOL+STZ (n=5). STZ was given a single dose, while mitoTEMPOL was given for 5 weeks after 1 week of STZ injection. Histopathological appearance, lipid droplets, mitophagy, and autophagy gene expression were examined after the mitoTEMPOL treatment. RESULTS We found metabolic zone shifting that might be correlated with the liver activity of fatty acid oxidation in the STZ group, a decrease of lipid droplets in mitoTEMPOL and mitoTEMPOL + STZ compared with Control and STZ groups were found in this study. We also found significant changes in PINK1, Parkin, BNIP3, Mfn1, and LC3 gene expression, but no difference in Opa1, Fis1, Drp1, and p62 gene expression, suggesting a change of mitochondrial fusion rather than mitochondrial fission correlated with mitophagy. CONCLUSION All this concluded that mitoTEMPOL could act as a modulator of mitophagy and metabolic function of the liver, thus amplifying its crucial role in preventing mitochondrial damage in the liver in the early onset of diabetes mellitus.
Collapse
Affiliation(s)
- Rova Virgana
- Department of Ophthalmology, Universitas Padjadjaran, Bandung, West Java, Indonesia,Cicendo National Eye Hospital, Bandung, West Java, Indonesia, These authors contributed eqully to this work
| | - Julia Windi Gunadi
- Department of Physiology, Faculty of Medicine, Maranatha Christian University, Bandung, West Java, Indonesia , These authors contributed eqully to this work,Corresponding author: Julia Windi Gunadi. Department of Physiology, Faculty of Medicine, Maranatha Christian University, Bandung, West Java, Indonesia. Tel: +622-2012186; Fax: +622-2015154;
| | - Nur Atik
- Biology Cell Division, Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, Indonesia
| | - Kwee Limdawati
- Department of Internal Medicine, Faculty of Medicine, Maranatha Christian University, Bandung, West Java, Indonesia
| | - Diana Krisanti Jasaputra
- Department of Pharmacology, Faculty of Medicine, Maranatha Christian University, Bandung, West Java, Indonesia
| | - Roro Wahyudianingsih
- Department of Pathology Anatomy, Faculty of Medicine, Maranatha Christian University, Bandung, West Java, Indonesia
| | | | | | - Hanna Goenawan
- Physiology Division, Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, Indonesia ,Physiology Molecular Laboratory, Biological Activity Division, Central Laboratory, Universitas Padjadjaran, Jatinangor, West Java, Indonesia
| | - Ronny Lesmana
- Physiology Division, Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, Indonesia ,Physiology Molecular Laboratory, Biological Activity Division, Central Laboratory, Universitas Padjadjaran, Jatinangor, West Java, Indonesia
| | - Arief Sjamsulaksan Kartasasmita
- Department of Ophthalmology, Universitas Padjadjaran, Bandung, West Java, Indonesia,Cicendo National Eye Hospital, Bandung, West Java, Indonesia
| |
Collapse
|
35
|
Abdullah CS, Remex NS, Aishwarya R, Nitu S, Kolluru GK, Traylor J, Hartman B, King J, Bhuiyan MAN, Hall N, Murnane KS, Goeders NE, Kevil CG, Orr AW, Bhuiyan MS. Mitochondrial dysfunction and autophagy activation are associated with cardiomyopathy developed by extended methamphetamine self-administration in rats. Redox Biol 2022; 58:102523. [PMID: 36335762 PMCID: PMC9641018 DOI: 10.1016/j.redox.2022.102523] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022] Open
Abstract
The recent rise in illicit use of methamphetamine (METH), a highly addictive psychostimulant, is a huge health care burden due to its central and peripheral toxic effects. Mounting clinical studies have noted that METH use in humans is associated with the development of cardiomyopathy; however, preclinical studies and animal models to dissect detailed molecular mechanisms of METH-associated cardiomyopathy development are scarce. The present study utilized a unique very long-access binge and crash procedure of METH self-administration to characterize the sequelae of pathological alterations that occur with METH-associated cardiomyopathy. Rats were allowed to intravenously self-administer METH for 96 h continuous weekly sessions over 8 weeks. Cardiac function, histochemistry, ultrastructure, and biochemical experiments were performed 24 h after the cessation of drug administration. Voluntary METH self-administration induced pathological cardiac remodeling as indicated by cardiomyocyte hypertrophy, myocyte disarray, interstitial and perivascular fibrosis accompanied by compromised cardiac systolic function. Ultrastructural examination and native gel electrophoresis revealed altered mitochondrial morphology and reduced mitochondrial oxidative phosphorylation (OXPHOS) supercomplexes (SCs) stability and assembly in METH exposed hearts. Redox-sensitive assays revealed significantly attenuated mitochondrial respiratory complex activities with a compensatory increase in pyruvate dehydrogenase (PDH) activity reminiscent of metabolic remodeling. Increased autophagy flux and increased mitochondrial antioxidant protein level was observed in METH exposed heart. Treatment with mitoTEMPO reduced the autophagy level indicating the involvement of mitochondrial dysfunction in the adaptive activation of autophagy in METH exposed hearts. Altogether, we have reported a novel METH-associated cardiomyopathy model using voluntary drug seeking behavior. Our studies indicated that METH self-administration profoundly affects mitochondrial ultrastructure, OXPHOS SCs assembly and redox activity accompanied by increased PDH activity that may underlie observed cardiac dysfunction.
Collapse
Affiliation(s)
- Chowdhury S Abdullah
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Naznin Sultana Remex
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Richa Aishwarya
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Sadia Nitu
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Gopi K Kolluru
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - James Traylor
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Brandon Hartman
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Judy King
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Mohammad Alfrad Nobel Bhuiyan
- Department of Medicine, Division of Clinical Informatics, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Nicole Hall
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Kevin Sean Murnane
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA; Department of Psychiatry, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Nicholas E Goeders
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Christopher G Kevil
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - A Wayne Orr
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Md Shenuarin Bhuiyan
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA.
| |
Collapse
|
36
|
Junghans M, John F, Cihankaya H, Schliebs D, Winklhofer KF, Bader V, Matschke J, Theiss C, Matschke V. ROS scavengers decrease γH2ax spots in motor neuronal nuclei of ALS model mice in vitro. Front Cell Neurosci 2022; 16:963169. [PMID: 36119129 PMCID: PMC9470831 DOI: 10.3389/fncel.2022.963169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/08/2022] [Indexed: 12/05/2022] Open
Abstract
Background: Amyotrophic lateral sclerosis (ALS) is an incurable neurodegenerative disease characterized by the loss of motor neurons in cerebral cortex, brainstem and spinal cord. Numerous studies have demonstrated signs of oxidative stress in postmortem neuronal tissue, cerebrospinal fluid, plasma and urine of ALS patients, without focusing on the specific processes within motor neurons. Thus, we aimed to investigate the relevance of reactive oxygen species (ROS) detoxification mechanisms and its consequences on the formation of toxic/lethal DNA double strand breaks (DSBs) in the ALS model of the Wobbler mouse. Methods: Live cell imaging in dissociated motor neuronal cultures was used to investigate the production of ROS using Dihydroethidium (DHE). The expression levels of ROS detoxifying molecules were investigated by qPCR as well as Western blots. Furthermore, the expression levels of DNA damage response proteins p53bp1 and H2ax were investigated using qPCR and immunofluorescence staining. Proof-of-principle experiments using ROS scavengers were performed in vitro to decipher the influence of ROS on the formation of DNA double strand breaks quantifying the γH2ax spots formation. Results: Here, we verified an elevated ROS-level in spinal motor neurons of symptomatic Wobbler mice in vitro. As a result, an increased number of DNA damage response proteins p53bp1 and γH2ax in dissociated motor neurons of the spinal cord of Wobbler mice was observed. Furthermore, we found a significantly altered expression of several antioxidant molecules in the spinal cord of Wobbler mice, suggesting a deficit in ROS detoxification mechanisms. This hypothesis could be verified by using ROS scavenger molecules in vitro to reduce the number of γH2ax foci in dissociated motor neurons and thus counteract the harmful effects of ROS. Conclusion: Our data indicate that maintenance of redox homeostasis may play a key role in the therapy of the neurodegenerative disease ALS. Our results underline a necessity for multimodal treatment approaches to prolong the average lifespan of motor neurons and thus slow down the progression of the disease, since a focused intervention in one pathomechanism seems to be insufficient in ALS therapy.
Collapse
Affiliation(s)
- Maya Junghans
- Department of Cytology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| | - Felix John
- Department of Cytology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| | - Hilal Cihankaya
- Department of Cytology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| | - Daniel Schliebs
- Department of Cytology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| | - Konstanze F. Winklhofer
- Department of Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
- Cluster of Excellence RESOLV, Bochum, Germany
| | - Verian Bader
- Department of Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
- Department of Biochemistry of Neurodegenerative Diseases, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| | - Johann Matschke
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Carsten Theiss
- Department of Cytology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| | - Veronika Matschke
- Department of Cytology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
- *Correspondence: Veronika Matschke
| |
Collapse
|
37
|
Adelusi OB, Ramachandran A, Lemasters JJ, Jaeschke H. The role of Iron in lipid peroxidation and protein nitration during acetaminophen-induced liver injury in mice. Toxicol Appl Pharmacol 2022; 445:116043. [PMID: 35513057 PMCID: PMC9843742 DOI: 10.1016/j.taap.2022.116043] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 01/19/2023]
Abstract
Acetaminophen (APAP) hepatotoxicity, a leading cause of acute liver failure in western countries, is characterized by mitochondrial superoxide and peroxynitrite formation. However, the role of iron, especially as facilitator of lipid peroxidation (LPO), has been controversial. Our aim was to determine the mechanism by which iron promotes cell death in this context. Fasted male C57BL/6J mice were treated with the iron chelator deferoxamine, minocycline (inhibitor of the mitochondrial calcium uniporter) or vehicle 1 h before 300 mg/kg APAP. Deferoxamine and minocycline significantly attenuated APAP-induced elevations in serum alanine amino transferase levels and hepatic necrosis at 6 h. This protection correlated with reduced 3-nitro-tyrosine protein adducts; LPO (malondialdehyde, 4-hydroxynonenal) was not detected. Activation of c-jun N-terminal kinase (JNK) was not affected but mitochondrial release of intermembrane proteins was reduced suggesting that the effect of iron was at the level of mitochondria. Co-treatment of APAP with FeSO4 exacerbated liver injury and protein nitration and triggered significant LPO; all effects were reversed by deferoxamine. Thus, after APAP overdose, iron imported into mitochondria facilitates protein nitration by peroxynitrite triggering mitochondrial dysfunction and cell death. Under these conditions, endogenous defense mechanisms largely prevent LPO. However, after iron overload, protein nitration and LPO contribute to APAP hepatotoxicity.
Collapse
Affiliation(s)
- Olamide B Adelusi
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - John J Lemasters
- Center for Cell Death, Injury & Regeneration, Departments of Drug Discovery & Biomedical Sciences and Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
38
|
Cai X, Cai H, Wang J, Yang Q, Guan J, Deng J, Chen Z. Molecular pathogenesis of acetaminophen-induced liver injury and its treatment options. J Zhejiang Univ Sci B 2022; 23:265-285. [PMID: 35403383 DOI: 10.1631/jzus.b2100977] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Acetaminophen, also known as N-acetyl-p-aminophenol (APAP), is commonly used as an antipyretic and analgesic agent. APAP overdose can induce hepatic toxicity, known as acetaminophen-induced liver injury (AILI). However, therapeutic doses of APAP can also induce AILI in patients with excessive alcohol intake or who are fasting. Hence, there is a need to understand the potential pathological mechanisms underlying AILI. In this review, we summarize three main mechanisms involved in the pathogenesis of AILI: hepatocyte necrosis, sterile inflammation, and hepatocyte regeneration. The relevant factors are elucidated and discussed. For instance, N-acetyl-p-benzoquinone imine (NAPQI) protein adducts trigger mitochondrial oxidative/nitrosative stress during hepatocyte necrosis, danger-associated molecular patterns (DAMPs) are released to elicit sterile inflammation, and certain growth factors contribute to liver regeneration. Finally, we describe the current potential treatment options for AILI patients and promising novel strategies available to researchers and pharmacists. This review provides a clearer understanding of AILI-related mechanisms to guide drug screening and selection for the clinical treatment of AILI patients in the future.
Collapse
Affiliation(s)
- Xiaopeng Cai
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Huiqiang Cai
- Department of Clinical Medicine, University of Aarhus, Palle Juul-Jensens Boulevard 82, 8200 Aarhus N, Denmark
| | - Jing Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Qin Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jun Guan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jingwen Deng
- Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China. , .,Department of Pathology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China. ,
| | - Zhi Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
39
|
Capacity of extracellular globins to reduce liver fibrosis via scavenging reactive oxygen species and promoting MMP-1 secretion. Redox Biol 2022; 52:102286. [PMID: 35334247 PMCID: PMC8956869 DOI: 10.1016/j.redox.2022.102286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/05/2022] [Accepted: 03/11/2022] [Indexed: 11/15/2022] Open
Abstract
Background & aims Hepatic stellate cells (HSCs) are the primary cell type in liver fibrosis, a significant global health care burden. Cytoglobin (CYGB), a globin family member expressed in HSCs, inhibits HSC activation and reduces collagen production. We studied the antifibrotic properties of globin family members hemoglobin (HB), myoglobin (MB), and neuroglobin (NGB) in comparison with CYGB. Approach & results We characterized the biological activities of globins in cultured human HSCs (HHSteCs) and their effects on carbon tetrachloride (CCl4)-induced cirrhosis in mice. All globins demonstrated greater antioxidant capacity than glutathione in cell-free systems. Cellular fractionation revealed endocytosis of extracellular MB, NGB, and CYGB, but not HB; endocytosed globins localized to intracellular membranous, cytoplasmic, and cytoskeletal fractions. MB, NGB, and CYGB, but not HB, scavenged reactive oxygen species generated spontaneously or stimulated by H2O2 or transforming growth factor β1 in HHSteCs and reduced collagen 1A1 production via suppressing COL1A1 promoter activity. Disulfide bond-mutant NGB displayed decreased heme and superoxide scavenging activity and reduced collagen inhibitory capacity. RNA sequencing of MB- and NGB-treated HHSteCs revealed downregulation of extracellular matrix–encoding and fibrosis-related genes and HSC deactivation markers. Upregulation of matrix metalloproteinase (MMP)-1 was observed following MB and NGB treatment, and MMP-1 knockdown partially reversed globin-mediated effects on secreted collagen. Importantly, administration of MB, NGB, and CYGB suppressed CCl4-induced mouse liver fibrosis. Conclusions These findings revealed unexpected roles for MB and NGB in deactivating HSCs and inhibiting liver fibrosis development, suggesting that globin therapy may represent a new strategy for combating fibrotic liver disease. Myoglobin, neuroglobin, and cytoglobin, but not hemoglobin:Internalize into human hepatic stellate cells via endocytosis pathway. Scavenge intracellular reactive oxidative species. Suppress COL1A1 promoter activity and promote matrix metaloproteinase-1 secretion. Suppress carbon tetrachloride-induced mouse liver fibrosis.
Collapse
|
40
|
Chen JW, Ma PW, Yuan H, Wang WL, Lu PH, Ding XR, Lun YQ, Yang Q, Lu LJ. mito-TEMPO Attenuates Oxidative Stress and Mitochondrial Dysfunction in Noise-Induced Hearing Loss via Maintaining TFAM-mtDNA Interaction and Mitochondrial Biogenesis. Front Cell Neurosci 2022; 16:803718. [PMID: 35210991 PMCID: PMC8861273 DOI: 10.3389/fncel.2022.803718] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 01/14/2022] [Indexed: 12/25/2022] Open
Abstract
The excessive generation of reactive oxygen species (ROS) and mitochondrial damage have been widely reported in noise-induced hearing loss (NIHL). However, the specific mechanism of noise-induced mitochondrial damage remains largely unclear. In this study, we showed that acoustic trauma caused oxidative damage to mitochondrial DNA (mtDNA), leading to the reduction of mtDNA content, mitochondrial gene expression and ATP level in rat cochleae. The expression level and mtDNA-binding function of mitochondrial transcription factor A (TFAM) were impaired following acoustic trauma without affecting the upstream PGC-1α and NRF-1. The mitochondria-target antioxidant mito-TEMPO (MT) was demonstrated to enter the inner ear after the systemic administration. MT treatment significantly alleviated noise-induced auditory threshold shifts 3d and 14d after noise exposure. Furthermore, MT significantly reduced outer hair cell (OHC) loss, cochlear ribbon synapse loss, and auditory nerve fiber (ANF) degeneration after the noise exposure. In addition, we found that MT treatment effectively attenuated noise-induced cochlear oxidative stress and mtDNA damage, as indicated by DHE, 4-HNE, and 8-OHdG. MT treatment also improved mitochondrial biogenesis, ATP generation, and TFAM-mtDNA interaction in the cochlea. These findings suggest that MT has protective effects against NIHL via maintaining TFAM-mtDNA interaction and mitochondrial biogenesis based on its ROS scavenging capacity.
Collapse
Affiliation(s)
- Jia-Wei Chen
- Department of Otolaryngology Head and Neck Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Peng-Wei Ma
- Department of Otolaryngology Head and Neck Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Hao Yuan
- Department of Otolaryngology Head and Neck Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Wei-Long Wang
- Department of Otolaryngology Head and Neck Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Pei-Heng Lu
- Department of Otolaryngology Head and Neck Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Xue-Rui Ding
- Department of Otolaryngology Head and Neck Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yu-Qiang Lun
- Department of Otolaryngology Head and Neck Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Qian Yang
- Department of Experimental Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Lian-Jun Lu
- Department of Otolaryngology Head and Neck Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
41
|
Yoshinaga N, Numata K. Rational Designs at the Forefront of Mitochondria-Targeted Gene Delivery: Recent Progress and Future Perspectives. ACS Biomater Sci Eng 2022; 8:348-359. [PMID: 34979085 DOI: 10.1021/acsbiomaterials.1c01114] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mitochondria play an essential role in cellular metabolism and generate energy in cells. To support these functions, several proteins are encoded in the mitochondrial DNA (mtDNA). The mutation of mtDNA causes mitochondrial dysfunction and ultimately results in a variety of inherited diseases. To date, gene delivery systems targeting mitochondria have been developed to ameliorate mtDNA mutations. However, applications of these strategies in mitochondrial gene therapy are still being explored and optimized. Thus, from this perspective, we herein highlight recent mitochondria-targeting strategies for gene therapy and discuss future directions for effective mitochondria-targeted gene delivery.
Collapse
Affiliation(s)
- Naoto Yoshinaga
- Biomacromolecule Research Team, RIKEN Center for Sustainable Resource Science, Saitama 351-0198, Japan
| | - Keiji Numata
- Biomacromolecule Research Team, RIKEN Center for Sustainable Resource Science, Saitama 351-0198, Japan.,Department of Material Chemistry, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
42
|
Shan S, Liu Z, Li L, Zhang C, Kou R, Song F. Calpain-mediated cleavage of mitochondrial fusion/fission proteins in acetaminophen-induced mice liver injury. Hum Exp Toxicol 2022; 41:9603271221108321. [PMID: 35713544 DOI: 10.1177/09603271221108321] [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: 11/16/2022]
Abstract
Mitochondrial dysfunction was considered to be a critical event in acetaminophen (APAP) -induced hepatotoxicity. Recent studies suggest that abnormal mitochondrial dynamics contributes to mitochondrial dysfunction in APAP-induced liver injury, yet the underlying mechanisms responsible for deregulated mitochondrial dynamics remains elusive. In this study, C57BL/6 mice were used to establish a model of acute liver injury via intraperitoneal (i.p.) injection with overdose of APAP. Furthermore, calpain intervention experiments were achieved by the inhibitors ALLN or calpeptin. The activity of serum enzymes and pathological changes of APAP-treated mice were evaluated, and the critical molecules in mitochondrial dynamics and calpain degradative pathway were determined by electron microscopy, immunoblot and calpain activity kit. The results demonstrated that APAP overdose resulted in a severe liver injury, mitochondrial damage and an obvious cleavage of fusion/fission proteins. Meanwhile, the activation of calpain degradative machinery in liver were observed following APAP. By contrast, pretreatment of calpain inhibitors significantly inhibited the activation of calpains. Our further investigation found that ALLN or calpeptin administration significantly suppresses the changes of mitochondrial dynamics in APAP-treated mice and finally protected against APAP-induced hepatoxicity. Overall, these results suggest that calpain-mediated cleavage of mitochondrial dynamics proteins was involved in the pathogenic process of mitochondrial dysfunction and thus present a potential molecular coupling APAP-induced hepatotoxicity.
Collapse
Affiliation(s)
- Shulin Shan
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, 12589Shandong University, Jinan, China
| | - Zhaoxiong Liu
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, 12589Shandong University, Jinan, China
| | - Linlin Li
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, 12589Shandong University, Jinan, China
| | - Cuiqin Zhang
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, 12589Shandong University, Jinan, China
| | - Ruirui Kou
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, 12589Shandong University, Jinan, China
| | - Fuyong Song
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, 12589Shandong University, Jinan, China
| |
Collapse
|
43
|
Jaeschke H, Adelusi OB, Akakpo JY, Nguyen NT, Sanchez-Guerrero G, Umbaugh DS, Ding WX, Ramachandran A. Recommendations for the use of the acetaminophen hepatotoxicity model for mechanistic studies and how to avoid common pitfalls. Acta Pharm Sin B 2021; 11:3740-3755. [PMID: 35024303 PMCID: PMC8727921 DOI: 10.1016/j.apsb.2021.09.023] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/22/2021] [Accepted: 09/10/2021] [Indexed: 02/07/2023] Open
Abstract
Acetaminophen (APAP) is a widely used analgesic and antipyretic drug, which is safe at therapeutic doses but can cause severe liver injury and even liver failure after overdoses. The mouse model of APAP hepatotoxicity recapitulates closely the human pathophysiology. As a result, this clinically relevant model is frequently used to study mechanisms of drug-induced liver injury and even more so to test potential therapeutic interventions. However, the complexity of the model requires a thorough understanding of the pathophysiology to obtain valid results and mechanistic information that is translatable to the clinic. However, many studies using this model are flawed, which jeopardizes the scientific and clinical relevance. The purpose of this review is to provide a framework of the model where mechanistically sound and clinically relevant data can be obtained. The discussion provides insight into the injury mechanisms and how to study it including the critical roles of drug metabolism, mitochondrial dysfunction, necrotic cell death, autophagy and the sterile inflammatory response. In addition, the most frequently made mistakes when using this model are discussed. Thus, considering these recommendations when studying APAP hepatotoxicity will facilitate the discovery of more clinically relevant interventions.
Collapse
Key Words
- AIF, apoptosis-inducing factor
- AMPK, AMP-activated protein kinase
- APAP, acetaminophen
- ARE, antioxidant response element
- ATG, autophagy-related genes
- Acetaminophen hepatotoxicity
- Apoptosis
- Autophagy
- BSO, buthionine sulfoximine
- CAD, caspase-activated DNase
- CYP, cytochrome P450 enzymes
- DAMPs, damage-associated molecular patterns
- DMSO, dimethylsulfoxide
- Drug metabolism
- EndoG, endonuclease G
- FSP1, ferroptosis suppressing protein 1
- Ferroptosis
- GPX4, glutathione peroxidase 4
- GSH, glutathione
- GSSG, glutathione disulfide
- Gclc, glutamate–cysteine ligase catalytic subunit
- Gclm, glutamate–cysteine ligase modifier subunit
- HMGB1, high mobility group box protein 1
- HNE, 4-hydroxynonenal
- Innate immunity
- JNK, c-jun N-terminal kinase
- KEAP1, Kelch-like ECH-associated protein 1
- LAMP, lysosomal-associated membrane protein
- LC3, light chain 3
- LOOH, lipid hydroperoxides
- LPO, lipid peroxidation
- MAP kinase, mitogen activated protein kinase
- MCP-1, monocyte chemoattractant protein-1
- MDA, malondialdehyde
- MPT, mitochondrial permeability transition
- Mitochondria
- MnSOD, manganese superoxide dismutase
- NAC, N-acetylcysteine
- NAPQI, N-acetyl-p-benzoquinone imine
- NF-κB, nuclear factor κB
- NQO1, NAD(P)H:quinone oxidoreductase 1
- NRF2
- NRF2, nuclear factor erythroid 2-related factor 2
- PUFAs, polyunsaturated fatty acids
- ROS, reactive oxygen species
- SMAC/DIABLO, second mitochondria-derived activator of caspase/direct inhibitor of apoptosis-binding protein with low pI
- TLR, toll like receptor
- TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling
- UGT, UDP-glucuronosyltransferases
- mTORC1, mammalian target of rapamycin complex 1
Collapse
Affiliation(s)
- Hartmut Jaeschke
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Olamide B. Adelusi
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jephte Y. Akakpo
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Nga T. Nguyen
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Giselle Sanchez-Guerrero
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - David S. Umbaugh
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
44
|
Matsuoka Y, Takahashi M, Sugiura Y, Izumi Y, Nishiyama K, Nishida M, Suematsu M, Bamba T, Yamada KI. Structural library and visualization of endogenously oxidized phosphatidylcholines using mass spectrometry-based techniques. Nat Commun 2021; 12:6339. [PMID: 34732715 PMCID: PMC8566498 DOI: 10.1038/s41467-021-26633-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 10/14/2021] [Indexed: 11/09/2022] Open
Abstract
Although oxidized phosphatidylcholines (oxPCs) play critical roles in numerous pathological events, the type and production sites of endogenous oxPCs remain unknown because of the lack of structural information and dedicated analytical methods. Herein, a library of 465 oxPCs is constructed using high-resolution mass spectrometry-based non-targeted analytical methods and employed to detect 70 oxPCs in mice with acetaminophen-induced acute liver failure. We show that doubly oxygenated polyunsaturated fatty acid (PUFA)-PCs (PC PUFA;O2), containing epoxy and hydroxide groups, are generated in the early phase of liver injury. Hybridization with in-vivo 18O labeling and matrix-assisted laser desorption/ionization-tandem MS imaging reveals that PC PUFA;O2 are accumulated in cytochrome P450 2E1-expressing and glutathione-depleted hepatocytes, which are the major sites of liver injury. The developed library and visualization methodology should facilitate the characterization of specific lipid peroxidation events and enhance our understanding of their physiological and pathological significance in lipid peroxidation-related diseases.
Collapse
Affiliation(s)
- Yuta Matsuoka
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Masatomo Takahashi
- Metabolomics Laboratory, Research Center for Transomics Medicine, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuki Sugiura
- Department of Biochemistry, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yoshihiro Izumi
- Metabolomics Laboratory, Research Center for Transomics Medicine, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kazuhiro Nishiyama
- Department of Physiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Motohiro Nishida
- Department of Physiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan.,Division of Cardiocirculatory Signaling, National Institute for Physiological Sciences and Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, 444-8787, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Takeshi Bamba
- Metabolomics Laboratory, Research Center for Transomics Medicine, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Ken-Ichi Yamada
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
45
|
Zarei F, Daghigh-Kia H, Masoudi R. Supplementation of ram's semen extender with Mito-TEMPO II: Quality evaluation and flow cytometry study of post-thawed spermatozoa. Andrologia 2021; 54:e14299. [PMID: 34730242 DOI: 10.1111/and.14299] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 09/27/2021] [Accepted: 10/25/2021] [Indexed: 11/30/2022] Open
Abstract
Cryopreservation is an effective method to spread qualified ram spermatozoa for reproductive goals in different farms, but cryopreservation's shocks reduce sperm quality. This study investigated the efficacy of the new mitochondria-targeted antioxidant Mito-TEMPO on post-thawed quality of spermatozoa in sheep. Collected samples were divided into five groups and after dilution, received different doses of Mito-TEMPO (0, 0.5, 5, 50 and 500 µM), and frozen. Thawed sperm motility parameters, malondialdehyde content, membrane functionality, abnormal morphology, mitochondria activity, acrosome integrity, DNA fragmentation, ROS concentration, viability and apoptotic-like changes, were evaluated. According to the results, Mito-TEMPO (5 and 50 μM) improved (p ≤ 0.05) motility parameters, average path velocity, membrane functionality, mitochondria activity and viability compared with the other groups. Moreover, apoptotic-like changes, lipid peroxidation and ROS concentration were lower (p ≤ 0.05) in groups received 5 and 50 μM Mito-TEMPO. Mito-TEMPO showed no effect (p > 0.05) on sperm acrosome integrity, morphology and DNA fragmentation. In conclusion, Mito-TEMPO as a targeted antioxidant could be an efficient cryo-additive to enhance quality parameters of post-thawed ram semen.
Collapse
Affiliation(s)
- Fateme Zarei
- Department of Animal Science, College of Agriculture, University of Tabriz, Tabriz, Iran
| | - Hossein Daghigh-Kia
- Department of Animal Science, College of Agriculture, University of Tabriz, Tabriz, Iran
| | - Reza Masoudi
- Animal Science Research Institute of Iran (ASRI), Agricultural Research Education and Extension Organization (AREEO), Karaj, Iran
| |
Collapse
|
46
|
Dobrinskikh E, Al-Juboori SI, Zarate MA, Zheng L, De Dios R, Balasubramaniyan D, Sherlock LG, Orlicky DJ, Wright CJ. Pulmonary implications of acetaminophen exposures independent of hepatic toxicity. Am J Physiol Lung Cell Mol Physiol 2021; 321:L941-L953. [PMID: 34585971 PMCID: PMC8616618 DOI: 10.1152/ajplung.00234.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 11/22/2022] Open
Abstract
Both preclinical and clinical studies have demonstrated that exposures to acetaminophen (APAP) at levels that cause hepatic injury cause pulmonary injury as well. However, whether exposures that do not result in hepatic injury have acute pulmonary implications is unknown. Thus, we sought to determine how APAP exposures at levels that do not result in significant hepatic injury impact the mature lung. Adult male ICR mice (8-12 wk) were exposed to a dose of APAP known to cause hepatotoxicity in adult mice [280 mg/kg, intraperitoneal (ip)], as well as a lower dose previously reported to not cause hepatic injury (140 mg/kg, ip). We confirm that the lower dose exposures did not result in significant hepatic injury. However, like high dose, lower exposure resulted in increased cellular content of the bronchoalveolar lavage fluid and induced a proinflammatory pulmonary transcriptome. Both the lower and higher dose exposures resulted in measurable changes in lung morphometrics, with the lower dose exposure causing alveolar wall thinning. Using RNAScope, we were able to detect dose-dependent, APAP-induced pulmonary Cyp2e1 expression. Finally, using FLIM we determined that both APAP exposures resulted in acute pulmonary metabolic changes consistent with mitochondrial overload in lower doses and a shift to glycolysis at a high dose. Our findings demonstrate that APAP exposures that do not cause significant hepatic injury result in acute inflammatory, morphometric, and metabolic changes in the mature lung. These previously unreported findings may help explain the potential relationship between APAP exposures and pulmonary-related morbidity.
Collapse
Affiliation(s)
- Evgenia Dobrinskikh
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Saif I Al-Juboori
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Miguel A Zarate
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Lijun Zheng
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Robyn De Dios
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Durga Balasubramaniyan
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Laura G Sherlock
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - David J Orlicky
- Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado
| | - Clyde J Wright
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
47
|
Hepatocyte-Specific Deficiency of BAP31 Amplified Acetaminophen-Induced Hepatotoxicity via Attenuating Nrf2 Signaling Activation in Mice. Int J Mol Sci 2021; 22:ijms221910788. [PMID: 34639126 PMCID: PMC8509202 DOI: 10.3390/ijms221910788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 01/18/2023] Open
Abstract
Liver-specific deficiency of B-cell receptor-associated protein 31 knockout mice (BAP31-LKO) and the littermates were injected with acetaminophen (APAP), markers of liver injury, and the potential molecular mechanisms were determined. In response to APAP overdose, serum aspartate aminotransferase and alanine aminotransferase levels were increased in BAP31-LKO mice than in wild-type controls, accompanied by enhanced liver necrosis. APAP-induced apoptosis and mortality were increased. Hepatic glutathione was decreased (1.60 ± 0.31 μmol/g tissue in WT mice vs. 0.85 ± 0.14 μmol/g tissue in BAP31-LKO mice at 6 h, p < 0.05), along with reduced glutathione reductase activity and superoxide dismutase; while malondialdehyde was significantly induced (0.41 ± 0.03 nmol/mg tissue in WT mice vs. 0.50 ± 0.05 nmol/mg tissue in BAP31-LKO mice for 6 h, p < 0.05). JNK signaling activation and APAP-induced hepatic inflammation were increased in BAP31-LKO mice. The mechanism research revealed that BAP31-deficiency decreased Nrf2 mRNA stability (half-life of Nrf2 mRNA decreased from ~1.3 h to ~40 min) and miR-223 expression, led to reduced nuclear factor erythroid 2-related factor 2 (Nrf2) signaling activation and antioxidant genes induction. BAP31-deficiency decreased mitochondrial membrane potentials, reduced mitochondria-related genes expression, and resulted in mitochondrial dysfunction in the liver. Conclusions: BAP31-deficiency reduced the antioxidant response and Nrf2 signaling activation via reducing Nrf2 mRNA stabilization, enhanced JNK signaling activation, hepatic inflammation, and apoptosis, amplified APAP-induced hepatotoxicity in mice.
Collapse
|
48
|
Esmaeilkhanian S, Asadzadeh N, Masoudi R. Flow cytometry study of post-thawed bulk spermatozoa: Mito-TEMPO improves cryopreservation performance by controlling apoptosis rate, DNA fragmentation and ROS production. Cryobiology 2021; 103:147-149. [PMID: 34562474 DOI: 10.1016/j.cryobiol.2021.09.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 09/20/2021] [Indexed: 11/24/2022]
Abstract
Sperm cryopreservation is used to spread qualified semen for artificial insemination, but the freezing process reduces sperm quality. This study assessed the efficacy of Mito-TEMPO on post-thawed goat sperm quality. Semen samples divided to five equal groups and after dilution, received different doses of Mito-TEMPO (0, 1, 10, 100 and 1000 μM), and cryopreserved in liquid nitrogen. After thawing, flow cytometry analysis was performed to evaluate sperm mitochondria membrane potential, viability, apoptotic-like changes, DNA fragmentation and ROS concentration. According to the results, Mito-TEMPO (10 and 100 μM) improved (P ≤ 0.05) sperm viability and decreased (P ≤ 0.05) apoptotic-like changes and ROS concentration compared to the other groups. Mitochondria membrane potential was higher (P ≤ 0.05) in groups received 1, 10 and 100 μM Mito-TEMPO. The lowest (P ≤ 0.05) DNA fragmentation was observed in group received 10 μM Mito-TEMPO. In conclusion, mitochondria-targeted antioxidant Mito-TEMPO could be an efficient cryo-additive to enhance flowcytometric quality parameters of post-thawed bulk semen.
Collapse
Affiliation(s)
- S Esmaeilkhanian
- Animal Science Research Institute of Iran (ASRI), Agricultural Research Education and Extension Organization (AREEO), Karaj, Iran
| | - N Asadzadeh
- Animal Science Research Institute of Iran (ASRI), Agricultural Research Education and Extension Organization (AREEO), Karaj, Iran
| | - R Masoudi
- Animal Science Research Institute of Iran (ASRI), Agricultural Research Education and Extension Organization (AREEO), Karaj, Iran.
| |
Collapse
|
49
|
Ramachandran A, Jaeschke H. Oxidant Stress and Acetaminophen Hepatotoxicity: Mechanism-Based Drug Development. Antioxid Redox Signal 2021; 35:718-733. [PMID: 34232786 PMCID: PMC8558076 DOI: 10.1089/ars.2021.0102] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Acetaminophen (APAP) is one of the quantitively most consumed drugs worldwide. Although safe at therapeutic doses, intentional or unintentional overdosing occurs frequently causing severe liver injury and even liver failure. In the United States, 50% of all acute liver failure cases are caused by APAP overdose. However, only one antidote with a limited therapeutic window, N-acetylcysteine, is clinically approved. Thus, more effective therapeutic interventions are urgently needed. Recent Advances: Although APAP hepatotoxicity has been extensively studied for almost 50 years, particular progress has been made recently in two areas. First, there is now a detailed understanding of involvement of oxidative and nitrosative stress in the pathophysiology, with identification of the reactive species involved, their initial generation in mitochondria, amplification through the c-Jun N-terminal kinase pathway, and the mechanisms of cell death. Second, it was demonstrated in human hepatocytes and through biomarkers in vivo that the mechanisms of liver injury in animals accurately reflect the human pathophysiology, which allows the translation of therapeutic targets identified in animals to patients. Critical Issues: For progress, solid understanding of the pathophysiology of APAP hepatotoxicity and of a drug's targets is needed to identify promising new therapeutic intervention strategies and drugs, which may be applied to humans. Future Directions: In addition to further refine the mechanistic understanding of APAP hepatotoxicity and identify additional drugs with complementary mechanisms of action to prevent cell death, more insight into the mechanisms of regeneration and developing of drugs, which promote recovery, remains a future challenge. Antioxid. Redox Signal. 35, 718-733.
Collapse
Affiliation(s)
- Anup Ramachandran
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
50
|
Morán-Costoya A, Proenza AM, Gianotti M, Lladó I, Valle A. Sex Differences in Nonalcoholic Fatty Liver Disease: Estrogen Influence on the Liver-Adipose Tissue Crosstalk. Antioxid Redox Signal 2021; 35:753-774. [PMID: 33736456 DOI: 10.1089/ars.2021.0044] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Significance: Nonalcoholic fatty liver disease (NAFLD) is a hepatic and systemic disorder with a complex multifactorial pathogenesis. Owing to the rising incidence of obesity and diabetes mellitus, the prevalence of NAFLD and its impact on global health care are expected to increase in the future. Differences in NAFLD exist between males and females, and among females depending on their reproductive status. Clinical and preclinical data show that females in the fertile age are more protected against NAFLD, and studies in postmenopausal women and ovariectomized animal models support a protective role for estrogens. Recent Advances: An efficient crosstalk between the liver and adipose tissue is necessary to regulate lipid and glucose metabolism, protecting the liver from steatosis and insulin resistance contributing to NALFD. New advances in the knowledge of sexual dimorphism in liver and adipose tissue are providing interesting clues about the sex differences in NAFLD pathogenesis that could inspire new therapeutic strategies. Critical Issues: Sex hormones influence key master regulators of lipid metabolism and oxidative stress in liver and adipose tissue. All these sex-biased metabolic adjustments shape the crosstalk between liver and adipose tissue, contributing to the higher protection of females to NAFLD. Future Directions: The development of novel drugs based on the protective action of estrogens, but without its feminizing or undesired side effects, might provide new therapeutic strategies for the management of NAFLD. Antioxid. Redox Signal. 35, 753-774.
Collapse
Affiliation(s)
- Andrea Morán-Costoya
- Energy Metabolism and Nutrition Group, Department of Fundamental Biology and Health Sciences, Research Institute of Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Ana M Proenza
- Energy Metabolism and Nutrition Group, Department of Fundamental Biology and Health Sciences, Research Institute of Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.,Center for Biomedical Research in the Pathophysiology of Obesity and Nutrition Network, Carlos III Health Institute, Madrid, Spain
| | - Magdalena Gianotti
- Energy Metabolism and Nutrition Group, Department of Fundamental Biology and Health Sciences, Research Institute of Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.,Center for Biomedical Research in the Pathophysiology of Obesity and Nutrition Network, Carlos III Health Institute, Madrid, Spain
| | - Isabel Lladó
- Energy Metabolism and Nutrition Group, Department of Fundamental Biology and Health Sciences, Research Institute of Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.,Center for Biomedical Research in the Pathophysiology of Obesity and Nutrition Network, Carlos III Health Institute, Madrid, Spain
| | - Adamo Valle
- Energy Metabolism and Nutrition Group, Department of Fundamental Biology and Health Sciences, Research Institute of Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.,Center for Biomedical Research in the Pathophysiology of Obesity and Nutrition Network, Carlos III Health Institute, Madrid, Spain
| |
Collapse
|