1
|
Wang X, Ma C, Mi K, Cao X, Tan Y, Yuan H, Ren J, Liang X. Urolithin A attenuates Doxorubicin-induced cardiotoxicity by enhancing PINK1-regulated mitophagy via Ambra1. Chem Biol Interact 2025; 406:111363. [PMID: 39725191 DOI: 10.1016/j.cbi.2024.111363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/02/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
Doxorubicin (Dox) is a widely used antineoplastics although its clinical usage is greatly limited by its cardiotoxicity. Several studies have depicted an essential role for dampened mitophagy and mitochondrial injury in Dox cardiotoxicity. However, preventative measure to alleviate Dox-evoked cardiotoxicity via targeting mitophagy and mitochondrial integrity remains elusive. Urolithin A (UA) is a newly identified mitophagy inducer with antioxidant and anti-apoptotic properties although its effect on Dox-induced cardiotoxicity is unknown. This study was designed to explore the effect of UA on Dox cardiotoxicity and mechanisms involved. Our results indicated that UA alleviated Dox-induced cardiac dysfunction exhibited by echocardiographic parameters and histological analyses, and partially relieved Dox-induced apoptosis in vitro and in vivo, and mitochondrial dysfunction including ΔΨm dissipation and ROS production in vitro. The ability of UA to facilitate restoration of mitophagy in mice and H9C2s underscored its advantageous effects, manifested as upregulation of mitophagy-related proteins, including p62, LC3, PINK1 and Parkin, as well as the co-location between LC3 and mitochondria. Incubation with 3 -MA nearly reversed the UA-evoked rise of mitophagy-related proteins, and inhibition of apoptosis. Given that knockdown of Ambra1 almost abolished UA-induced protective effect, the enhanced expression of Ambra1 owing to UA increased PINK1 levels by inhibiting its degradation via LONP1. Collectively, our results suggest that the cardioprotective properties of UA depend on the stimulation of PINK1-dependent mitophagy through promoting Ambra1 expression to inhibit PINK1 degradation by LONP1. This highlights UA's potential as a valuable treatment option and its importance in cardioprotective strategies against Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
| | - Chao Ma
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
| | - Keying Mi
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
| | - Xinran Cao
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China; Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
| | - Yinghua Tan
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
| | - Haitao Yuan
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China; Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China.
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
| | - Xinyue Liang
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China; Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China.
| |
Collapse
|
2
|
Sommerfeld-Klatta K, Jiers W, Rzepczyk S, Nowicki F, Łukasik-Głębocka M, Świderski P, Zielińska-Psuja B, Żaba Z, Żaba C. The Effect of Neuropsychiatric Drugs on the Oxidation-Reduction Balance in Therapy. Int J Mol Sci 2024; 25:7304. [PMID: 39000411 PMCID: PMC11242277 DOI: 10.3390/ijms25137304] [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: 04/21/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/16/2024] Open
Abstract
The effectiveness of available neuropsychiatric drugs in the era of an increasing number of patients is not sufficient, and the complexity of neuropsychiatric disease entities that are difficult to diagnose and therapeutically is increasing. Also, discoveries about the pathophysiology of neuropsychiatric diseases are promising, including those initiating a new round of innovations in the role of oxidative stress in the etiology of neuropsychiatric diseases. Oxidative stress is highly related to mental disorders, in the treatment of which the most frequently used are first- and second-generation antipsychotics, mood stabilizers, and antidepressants. Literature reports on the effect of neuropsychiatric drugs on oxidative stress are divergent. They are starting with those proving their protective effect and ending with those confirming disturbances in the oxidation-reduction balance. The presented publication reviews the state of knowledge on the role of oxidative stress in the most frequently used therapies for neuropsychiatric diseases using first- and second-generation antipsychotic drugs, i.e., haloperidol, clozapine, risperidone, olanzapine, quetiapine, or aripiprazole, mood stabilizers: lithium, carbamazepine, valproic acid, oxcarbazepine, and antidepressants: citalopram, sertraline, and venlafaxine, along with a brief pharmacological characteristic, preclinical and clinical studies effects.
Collapse
Affiliation(s)
- Karina Sommerfeld-Klatta
- Department of Toxicology, Poznań University of Medical Sciences, 3 Rokietnicka Street, 60-806 Poznań, Poland
| | - Wiktoria Jiers
- Department of Toxicology, Poznań University of Medical Sciences, 3 Rokietnicka Street, 60-806 Poznań, Poland
| | - Szymon Rzepczyk
- Department of Forensic Medicine, Poznań University of Medical Sciences, 10 Rokietnicka Street, 60-806 Poznań, Poland
| | - Filip Nowicki
- Department of Forensic Medicine, Poznań University of Medical Sciences, 10 Rokietnicka Street, 60-806 Poznań, Poland
| | - Magdalena Łukasik-Głębocka
- Department of Emergency Medicine, Poznań University of Medical Sciences, 7 Rokietnicka Street, 60-806 Poznań, Poland
| | - Paweł Świderski
- Department of Forensic Medicine, Poznań University of Medical Sciences, 10 Rokietnicka Street, 60-806 Poznań, Poland
| | - Barbara Zielińska-Psuja
- Department of Toxicology, Poznań University of Medical Sciences, 3 Rokietnicka Street, 60-806 Poznań, Poland
| | - Zbigniew Żaba
- Department of Emergency Medicine, Poznań University of Medical Sciences, 7 Rokietnicka Street, 60-806 Poznań, Poland
| | - Czesław Żaba
- Department of Forensic Medicine, Poznań University of Medical Sciences, 10 Rokietnicka Street, 60-806 Poznań, Poland
| |
Collapse
|
3
|
Robichaud K, Bragg LM, Servos MR, Craig PM. Venlafaxine exposure alters mitochondrial respiration and mitomiR abundance in zebrafish brains. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2024; 43:1569-1582. [PMID: 38695684 DOI: 10.1002/etc.5884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/14/2024] [Accepted: 04/04/2024] [Indexed: 06/27/2024]
Abstract
Wastewater treatment plant (WWTP) effluent often releases pharmaceuticals like venlafaxine (a serotonin-norephinephrine reuptake inhibitor antidepressant) to freshwater ecosystems at levels causing adverse metabolic effects on fish. Changes to fish metabolism can be regulated by epigenetic mechanisms like microRNA (small RNA molecules that regulate mRNA translation), including regulating mitochondrial mRNAs. Nuclear-encoded microRNAs regulate mitochondrial gene expression in mammals, and have predicted effects in fish. We aimed to identify whether venlafaxine exposure changed mitochondrial respiration and resulted in differentially abundant mitochondrial microRNA (mitomiRs) in zebrafish brains. In vitro exposure of brain homogenate to below environmentally relevant concentrations of venlafaxine (<1 µg/L) caused a decrease in mitochondrial respiration, although this was not driven by changes to mitochondrial Complex I or II function. To identify whether these effects occur in vivo, zebrafish were exposed to 1 µg/L venlafaxine for 0, 1, 6, 12, 24, and 96 h. In vivo, venlafaxine exposure had no significant effects on brain mitochondrial respiration; however, select mitomiRs (dre-miR-301a-5p, dre-miR-301b-3p, and dre-miR-301c-3p) were also measured, because they were bioinformatically predicted to regulate mitochondrial cytochrome c oxidase subunit I (COI) abundance. These mitomiRs were differentially regulated based on venlafaxine exposure (with miR-301c-3p abundance differing during the day and miR-301b-3p being lower in exposed fish at night), and with respect to sex and time sampled. Overall, the results demonstrated that in vitro venlafaxine exposure to zebrafish brain caused a decrease in mitochondrial respiration, but these effects were not seen after acute in vivo exposure. Results may have differed because in vivo exposure allows for fish to mitigate effects through mechanisms that could include mitomiR regulation, and because fish were only acutely exposed. Environ Toxicol Chem 2024;43:1569-1582. © 2024 The Authors. Environmental Toxicology and Chemistry published by Wiley Periodicals LLC on behalf of SETAC.
Collapse
Affiliation(s)
- Karyn Robichaud
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Leslie M Bragg
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Mark R Servos
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Paul M Craig
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
4
|
Migheli R, Lostia G, Galleri G, Rocchitta G, Serra PA, Campesi I, Bassareo V, Acquas E, Peana AT. New perspective for an old drug: Can naloxone be considered an antioxidant agent? Biochem Biophys Rep 2023; 34:101441. [PMID: 36875795 PMCID: PMC9975616 DOI: 10.1016/j.bbrep.2023.101441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/30/2023] [Accepted: 02/13/2023] [Indexed: 02/22/2023] Open
Abstract
Background Experimental evidence indicates that Naloxone (NLX) holds antioxidant properties. The present study aims at verifying the hypothesis that NLX could prevent oxidative stress induced by hydrogen peroxide (H2O2) in PC12 cells. Methods To investigate the antioxidant effect of NLX, initially, we performed electrochemical experiments by means of platinum-based sensors in a cell-free system. Subsequently, NLX was tested in PC12 cells on H2O2-induced overproduction of intracellular levels of reactive-oxygen-species (ROS), apoptosis, modification of cells' cycle distribution and damage of cells' plasma membrane. Results This study reveals that NLX counteracts intracellular ROS production, reduces H2O2-induced apoptosis levels, and prevents the oxidative damage-dependent increases of the percentage of cells in G2/M phase. Likewise, NLX protects PC12 cells from H2O2- induced oxidative damage, by preventing the lactate dehydrogenase (LDH) release. Moreover, electrochemical experiments confirmed the antioxidant properties of NLX. Conclusion Overall, these findings provide a starting point for studying further the protective effects of NLX on oxidative stress.
Collapse
Affiliation(s)
- Rossana Migheli
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100, Sassari, Italy
| | - Giada Lostia
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100, Sassari, Italy
| | - Grazia Galleri
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100, Sassari, Italy
| | - Gaia Rocchitta
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100, Sassari, Italy
| | - Pier Andrea Serra
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100, Sassari, Italy
| | - Ilaria Campesi
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy
| | - Valentina Bassareo
- Department of Biomedical Sciences and Center of Excellence for the Study of Neurobiology of Addiction, University of Cagliari, 09042, Monserrato, Cagliari, Italy
| | - Elio Acquas
- Department of Life and Environmental Sciences and Center of Excellence for the Study of Neurobiology of Addiction, University of Cagliari, 09042, Monserrato, Cagliari, Italy
| | - Alessandra T. Peana
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100, Sassari, Italy
| |
Collapse
|
5
|
Tang Y, Chen K, Xiao Z, Hong G, Hu L, Cai J, Lu Z. A novel mechanism of Dimethyl ester of Alpha-ketoglutarate in suppressing Paraquat-induced BEAS-2B cell injury by alleviating GSDME dependent pyroptosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 112:154698. [PMID: 36773430 DOI: 10.1016/j.phymed.2023.154698] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 01/09/2023] [Accepted: 02/02/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Acute lung injury (ALI) induced by paraquat (PQ) progresses rapidly, leading to high mortality; however, there is no specific antidote. Our limited knowledge of the pathogenic toxicological mechanisms of PQ has hindered the development of treatments against PQ exposure. PURPOSE Pyroptosis is a form of programmed cell death recently identified as a novel molecular mechanism adopted by chemotherapeutic drugs for cancer therapy. However, the involvement of pyroptosis in PQ-induced lung injury has not been reported. Therefore, we investigated the effects of PQ on the lung tissues to elucidate the molecular mechanisms underlying its toxicity, especially its ability to induce pyroptosis. METHODS To observe the morphological changes of BEAS-2B cells exposed to PQ, the plasma membrane damage of the cells was detected by LDH release assay, mitochondrial function and cell metabolism were detected by energy metabolism analysis. Western blotting was used to detect the protein levels of GSDMD, C-GSDMD, GSDME and N-GSDME in BEAS-2B cells. Metabolites of TCA cycle were detected by metabolomics, and the changes of TCA cycle metabolic enzymes in cells were detected by Western blotting. RESULTS We observed that PQ induced proteolytic cleavage of gasdermin E (GSDME) with concomitant cleavage of caspase 3 in BEAS-2B cells. Knockout of GSDME attenuated PQ-induced cell death. Additionally, PQ induced ROS accumulation, mitochondrial depolarisation, and mitochondrial dysfunction in these cells. PQ activated the caspase 3/GSDME pathway and damaged the cytoplasmic membrane in cells, leading to pyroptosis. We demonstrated that DMK suppressed PQ-induced pyroptosis by blocking PQ-induced caspase 3/GSDME pathway activation, reducing cellular ROS levels, and improving mitochondrial function. CONCLUSION These findings provide novel insights into the previously unrecognized mechanism of GSDME-dependent pyroptosis in PQ poisoning.
Collapse
Affiliation(s)
- Yahui Tang
- Department of Emergency Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Wenzhou Key Laboratory of emergency and disaster medicine, Wenzhou 325000, China.
| | - Kaiyuan Chen
- Department of Emergency Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Wenzhou Key Laboratory of emergency and disaster medicine, Wenzhou 325000, China
| | - Zhong Xiao
- Department of Emergency Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Wenzhou Key Laboratory of emergency and disaster medicine, Wenzhou 325000, China
| | - Guangliang Hong
- Department of Emergency Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Wenzhou Key Laboratory of emergency and disaster medicine, Wenzhou 325000, China
| | - Lufeng Hu
- Wenzhou Key Laboratory of emergency and disaster medicine, Wenzhou 325000, China; Department of Pharmacy, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jianping Cai
- Ministry of Health Key Laboratory of Geriatrics (J.-P.C.), Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, China
| | - Zhongqiu Lu
- Department of Emergency Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Wenzhou Key Laboratory of emergency and disaster medicine, Wenzhou 325000, China.
| |
Collapse
|
6
|
Meng H, Li R, Xie Y, Mo Z, Zhai H, Zhang G, Liang G, Shi X, Zhou B. Nanoparticles Mediated circROBO1 Silencing to Inhibit Hepatocellular Carcinoma Progression by Modulating miR-130a-5p/CCNT2 Axis. Int J Nanomedicine 2023; 18:1677-1693. [PMID: 37020690 PMCID: PMC10069521 DOI: 10.2147/ijn.s399318] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/07/2023] [Indexed: 03/31/2023] Open
Abstract
BACKGROUND Circular RNAs (circRNAs) are becoming vital biomarkers and therapeutic targets for malignant tumors due to their high stability and specificity in tissues. However, biological functions of circRNAs in hepatocellular carcinoma (HCC) are still not well studied. METHODS Gene Expression Omnibus (GEO) database and qRT-PCR were used to evaluate expression of circROBO1 (hsa_circ_0066568) in HCC tissues and cell lines. CCK-8, colony formation, EdU staining, flow cytometry for cell cycle analysis, and xenograft model assays were performed to detect the circROBO1 function in vitro and in vivo. RNA pull-down, RNA immunoprecipitation (RIP), and Luciferase reporter assays were used to investigate the relationship among circROBO1, miR-130a-5p, and CCNT2. More importantly, we developed nanoparticles made from poly lactic-co-glycolic acid (PLGA) and polyethylene glycol (PEG) chains as the delivery system of si-circROBO1 and then applied them to HCC in vitro and in mice. RESULTS circROBO1 was obviously upregulated in HCC tissues and cell lines, and elevated circROBO1 was closely correlated with worse prognosis for HCC patients. Functionally, knocking down circROBO1 significantly suppressed HCC cells growth in vitro and in mice. Mechanistically, circROBO1 acted as a competing endogenous RNA to downregulate miR-130a-5p, leading to CCNT2 expression upregulation. Furthermore, miR-130a-5p mimic or CCNT2 knockdown reversed the role of circROBO1 overexpression on HCC cells, which demonstrated that circROBO1 promoted HCC development via miR-130a-5p/CCNT2 axis. In addition, we developed nanoparticles loaded with si-circROBO1, named as PLGA-PEG (si-circROBO1) NPs, which significantly prevented the proliferation of HCC cells, and did not exhibit apparent toxicity to major organs in vivo. CONCLUSION Our findings firstly demonstrate that circROBO1 overexpression promotes HCC progression by regulating miR-130a-5p/CCNT2 axis, which may serve as an effective nanotherapeutic target for HCC treatment.
Collapse
Affiliation(s)
- Hongyu Meng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, People’s Republic of China
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Ruixi Li
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, People’s Republic of China
| | - Yuankang Xie
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, People’s Republic of China
| | - Zhaohong Mo
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Hang Zhai
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Guangquan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, People’s Republic of China
| | - Guohui Liang
- School of Clinical Medicine, Henan University, Kaifeng, People’s Republic of China
| | - Xianjie Shi
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, People’s Republic of China
| | - Boxuan Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, People’s Republic of China
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| |
Collapse
|
7
|
Zhang H, Xia N, Tang T, Nie S, Zha L, Zhang M, Lv B, Lu Y, Jiao J, Li J, Cheng X. Cholesterol suppresses human iTreg differentiation and nTreg function through mitochondria-related mechanisms. J Transl Med 2023; 21:224. [PMID: 36973679 PMCID: PMC10045251 DOI: 10.1186/s12967-023-03896-z] [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: 10/13/2022] [Accepted: 01/16/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Both the crystalline and soluble forms of cholesterol increase macrophage secretion of interleukin 1β (IL-1β), aggravating the inflammatory response in atherosclerosis (AS). However, the link between cholesterol and regulatory T cells (Tregs) remains unclear. This study aimed to investigate the effect of cholesterol treatment on Tregs. METHODS Differentiation of induced Tregs (iTregs) was analyzed using flow cytometry. The expression of hypoxia-inducible factor-1a (HIF-1a) and its target genes was measured by western blotting and/or RT-qPCR. Two reporter jurkat cell lines were constructed by lentiviral transfection. Mitochondrial function and the structure of natural Tregs (nTregs) were determined by tetramethylrhodamine (TMRM) and mitoSOX staining, Seahorse assay, and electron microscopy. The immunoregulatory function of nTregs was determined by nTreg-macrophage co-culture assay and ELISA. RESULTS Cholesterol treatment suppressed iTreg differentiation and impaired nTreg function. Mechanistically, cholesterol induced the production of mitochondrial reactive oxygen species (mtROS) in naïve T cells, inhibiting the degradation of HIF-1α and unleashing its inhibitory effects on iTreg differentiation. Furthermore, cholesterol-induced mitochondrial oxidative damage impaired the immunosuppressive function of nTregs. Mixed lymphocyte reaction and nTreg-macrophage co-culture assays revealed that cholesterol treatment compromised the ability of nTregs to inhibit pro-inflammatory conventional T cell proliferation and promote the anti-inflammatory functions of macrophages. Finally, mitoTEMPO (MT), a specific mtROS scavenger, restored iTreg differentiation and protected nTreg from further deterioration. CONCLUSION Our findings suggest that cholesterol may aggravate inflammation within AS plaques by acting on both iTregs and nTregs, and that MT may be a promising anti-atherogenic drug.
Collapse
Affiliation(s)
- Huanzhi Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Ni Xia
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Tingting Tang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Shaofang Nie
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Lingfeng Zha
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Min Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Bingjie Lv
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Yuzhi Lu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Jiao Jiao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Jingyong Li
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China.
| |
Collapse
|
8
|
Chang J, Wu H, Wu J, Liu M, Zhang W, Hu Y, Zhang X, Xu J, Li L, Yu P, Zhu J. Constructing a novel mitochondrial-related gene signature for evaluating the tumor immune microenvironment and predicting survival in stomach adenocarcinoma. J Transl Med 2023; 21:191. [PMID: 36915111 PMCID: PMC10012538 DOI: 10.1186/s12967-023-04033-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/03/2023] [Indexed: 03/16/2023] Open
Abstract
BACKGROUND The incidence and mortality of gastric cancer ranks fifth and fourth worldwide among all malignancies, respectively. Accumulating evidences have revealed the close relationship between mitochondrial dysfunction and the initiation and progression of stomach cancer. However, rare prognostic models for mitochondrial-related gene risk have been built up in stomach cancer. METHODS In current study, the expression and prognostic value of mitochondrial-related genes in stomach adenocarcinoma (STAD) patients were systematically analyzed to establish a mitochondrial-related risk model based on available TCGA and GEO databases. The tumor microenvironment (TME), immune cell infiltration, tumor mutation burden, and drug sensitivity of gastric adenocarcinoma patients were also investigated using R language, GraphPad Prism 8 and online databases. RESULTS We established a mitochondrial-related risk prognostic model including NOX4, ALDH3A2, FKBP10 and MAOA and validated its predictive power. This risk model indicated that the immune cell infiltration in high-risk group was significantly different from that in the low-risk group. Besides, the risk score was closely related to TME signature genes and immune checkpoint molecules, suggesting that the immunosuppressive tumor microenvironment might lead to poor prognosis in high-risk groups. Moreover, TIDE analysis demonstrated that combined analysis of risk score and immune score, or stromal score, or microsatellite status could more effectively predict the benefit of immunotherapy in STAD patients with different stratifications. Finally, rapamycin, PD-0325901 and dasatinib were found to be more effective for patients in the high-risk group, whereas AZD7762, CEP-701 and methotrexate were predicted to be more effective for patients in the low-risk group. CONCLUSIONS Our results suggest that the mitochondrial-related risk model could be a reliable prognostic biomarker for personalized treatment of STAD patients.
Collapse
Affiliation(s)
- Jingjia Chang
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Hao Wu
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Jin Wu
- Department of Pathology, Laboratory of Translational Medicine Research, Deyang People's Hospital, Deyang, China.,Key Laboratory of Tumor Molecular Research of Deyang, Deyang, China.,Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Ming Liu
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Wentao Zhang
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Yanfen Hu
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Xintong Zhang
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Jing Xu
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Li Li
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Pengfei Yu
- Department of Gastrointestinal Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, 710032, Shaanxi, China.
| | - Jianjun Zhu
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
9
|
Dong M, Ding Y, Liu Y, Xu Z, Hong H, Sun H, Huang X, Yu X, Chen Q. Molecular insights of 2,6-dichlorobenzoquinone-induced cytotoxicity in zebrafish embryo: Activation of ROS-mediated cell cycle arrest and apoptosis. ENVIRONMENTAL TOXICOLOGY 2023; 38:694-700. [PMID: 36454668 DOI: 10.1002/tox.23721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/18/2022] [Accepted: 11/20/2022] [Indexed: 06/17/2023]
Abstract
2,6-dichloro-1,4-benzoquinone (2,6-DCBQ), as an emerging disinfection by-product, has been frequently detected in waters, posing potential health risk on public health. Although some studies have pointed out that 2,6-DCBQ exposure can induce cytotoxicity, limited information is available for underlying mechanism for 2,6-DCBQ-induced cytotoxicity. To explore this mechanism, we assessed the levels of reactive oxygen species (ROS), acridine orange (AO) staining, and the mRNA transcriptions of genes (Chk2, Cdk2, Ccna, Ccnb and Ccne) involved in cell-cycle and genes (p53, bax, bcl-2 and caspase 3) involved in apoptosis in zebrafish embryo, after exposed to different concentrations (10, 30, 60, 90 and 120 μg/L) of 2,6-DCBQ for 72 h. Our results indicated that 2,6-DCBQ exposure induced ROS generation and cell apoptosis, and disturbed the mRNA transcription of genes related to cell cycle and apoptosis in zebrafish embryo. Moreover, we also found that 30 ~ 60 μg/L 2,6-DCBQ is the important transition from cell-cycle arrest to cell apoptosis. These results provided novel insight into 2,6-DCBQ-induced cytotoxicity.
Collapse
Affiliation(s)
- Mingyue Dong
- College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, China
| | - Yan Ding
- College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, China
| | - Yingying Liu
- College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, China
| | - Zeqiong Xu
- College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, China
| | - Huachang Hong
- College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, China
| | - Hongjie Sun
- College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, China
| | - Xianfeng Huang
- National and Local Joint Engineering Research Center for Ecological Treatment Technology of Urban Water Pollution, College of Life and Environmental Science, Wenzhou University, Wenzhou, China
| | - Xinwei Yu
- Key Laboratory of Health Risk Factors for Seafood of Zhejiang Province, Zhoushan Municipal Center for Disease Control and Prevention, Zhoushan, China
| | - Qiang Chen
- Department of Environmental and Occupational Health, Jinhua Center for Disease Control and Prevention, Jinhua, People's Republic of China
| |
Collapse
|
10
|
Huang X, Han C, Zhong J, Hu J, Jin Y, Zhang Q, Luo W, Liu R, Ling F. Low expression of the dynamic network markers FOS/JUN in pre-deteriorated epithelial cells is associated with the progression of colorectal adenoma to carcinoma. J Transl Med 2023; 21:45. [PMID: 36698183 PMCID: PMC9875500 DOI: 10.1186/s12967-023-03890-5] [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: 11/24/2022] [Accepted: 01/17/2023] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Deterioration of normal intestinal epithelial cells is crucial for colorectal tumorigenesis. However, the process of epithelial cell deterioration and molecular networks that contribute to this process remain unclear. METHODS Single-cell data and clinical information were downloaded from the Gene Expression Omnibus (GEO) database. We used the recently proposed dynamic network biomarker (DNB) method to identify the critical stage of epithelial cell deterioration. Data analysis and visualization were performed using R and Cytoscape software. In addition, Single-Cell rEgulatory Network Inference and Clustering (SCENIC) analysis was used to identify potential transcription factors, and CellChat analysis was conducted to evaluate possible interactions among cell populations. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and gene set variation analysis (GSVA) analyses were also performed. RESULTS The trajectory of epithelial cell deterioration in adenoma to carcinoma progression was delineated, and the subpopulation of pre-deteriorated epithelial cells during colorectal cancer (CRC) initialization was identified at the single-cell level. Additionally, FOS/JUN were identified as biomarkers for pre-deteriorated epithelial cell subpopulations in CRC. Notably, FOS/JUN triggered low expression of P53-regulated downstream pro-apoptotic genes and high expression of anti-apoptotic genes through suppression of P53 expression, which in turn inhibited P53-induced apoptosis. Furthermore, malignant epithelial cells contributed to the progression of pre-deteriorated epithelial cells through the GDF signaling pathway. CONCLUSIONS We demonstrated the trajectory of epithelial cell deterioration and used DNB to characterize pre-deteriorated epithelial cells at the single-cell level. The expression of DNB-neighboring genes and cellular communication were triggered by DNB genes, which may be involved in epithelial cell deterioration. The DNB genes FOS/JUN provide new insights into early intervention in CRC.
Collapse
Affiliation(s)
- Xiaoqi Huang
- grid.79703.3a0000 0004 1764 3838Guangdong Key Laboratory of Fermentation and Enzyme Engineering, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006 China
| | - Chongyin Han
- grid.79703.3a0000 0004 1764 3838Guangdong Key Laboratory of Fermentation and Enzyme Engineering, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006 China
| | - Jiayuan Zhong
- grid.79703.3a0000 0004 1764 3838School of Mathematics, South China University of Technology, Guangzhou, 510641 China
| | - Jiaqi Hu
- grid.79703.3a0000 0004 1764 3838Guangdong Key Laboratory of Fermentation and Enzyme Engineering, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006 China
| | - Yabin Jin
- grid.452881.20000 0004 0604 5998Institute of Clinical Research, The First People’s Hospital of Foshan, Foshan, 528000 China
| | - Qinqin Zhang
- grid.79703.3a0000 0004 1764 3838Guangdong Key Laboratory of Fermentation and Enzyme Engineering, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006 China
| | - Wei Luo
- grid.452881.20000 0004 0604 5998Institute of Clinical Research, The First People’s Hospital of Foshan, Foshan, 528000 China
| | - Rui Liu
- grid.79703.3a0000 0004 1764 3838School of Mathematics, South China University of Technology, Guangzhou, 510641 China ,grid.513189.7Pazhou Lab, Guangzhou, 510330 China
| | - Fei Ling
- grid.79703.3a0000 0004 1764 3838Guangdong Key Laboratory of Fermentation and Enzyme Engineering, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006 China
| |
Collapse
|
11
|
Wan C, Liu XQ, Chen M, Ma HH, Wu GL, Qiao LJ, Cai YF, Zhang SJ. Tanshinone IIA ameliorates Aβ transendothelial transportation through SIRT1-mediated endoplasmic reticulum stress. J Transl Med 2023; 21:34. [PMID: 36670462 PMCID: PMC9854034 DOI: 10.1186/s12967-023-03889-y] [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: 10/16/2022] [Accepted: 01/12/2023] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND The disruption of blood-brain barrier (BBB), predominantly made up by brain microvascular endothelial cells (BMECs), is one of the characteristics of Alzheimer's disease (AD). Thus, improving BMEC function may be beneficial for AD treatment. Tanshinone IIA (Tan IIA) has been proved to ameliorate the cognitive dysfunction of AD. Herein, we explored how Tan IIA affected the function of BMECs in AD. METHODS Aβ1-42-treated brain-derived endothelium cells.3 (bEnd.3 cells) was employed for in vitro experiments. And we performed molecular docking and qPCR to determine the targeting molecule of Tan IIA on Sirtuins family. The APPswe/PSdE9 (APP/PS1) mice were applied to perform the in vivo experiments. Following the behavioral tests, protein expression was determined through western blot and immunofluorescence. The activities of oxidative stress-related enzymes were analyzed by biochemically kits. Nissl staining and thioflavin T staining were conducted to reflect the neurodegeneration and Aβ deposition respectively. RESULTS Molecular docking and qPCR results showed that Tan IIA mainly acted on Sirtuin1 (SIRT1) in Sirtuins family. The inhibitor of SIRT1 (EX527) was employed to further substantiate that Tan IIA could attenuate SIRT1-mediated endoplasmic reticulum stress (ER stress) in BMECs. Behavioral tests suggested that Tan IIA could improve the cognitive deficits in APP/PS1 mice. Tan IIA administration increased SIRT1 expression and alleviated ER stress in APP/PS1 mice. In addition, LRP1 expression was increased and RAGE expression was decreased after Tan IIA administration in both animals and cells. CONCLUSION Tan IIA could promote Aβ transportation by alleviating SIRT1-mediated ER stress in BMECs, which ameliorated cognitive deficits in APP/PS1 mice.
Collapse
Affiliation(s)
- Can Wan
- grid.411866.c0000 0000 8848 7685Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 510405 Guangzhou, China ,grid.413402.00000 0004 6068 0570Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, 510120 Guangzhou, China ,grid.9227.e0000000119573309Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055 Shenzhen, China
| | - Xiao-Qi Liu
- grid.411866.c0000 0000 8848 7685Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 510405 Guangzhou, China ,grid.413402.00000 0004 6068 0570Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, 510120 Guangzhou, China
| | - Mei Chen
- grid.411866.c0000 0000 8848 7685Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 510405 Guangzhou, China ,grid.413402.00000 0004 6068 0570Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, 510120 Guangzhou, China
| | - Hui-Han Ma
- grid.411866.c0000 0000 8848 7685Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 510405 Guangzhou, China ,grid.413402.00000 0004 6068 0570Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, 510120 Guangzhou, China
| | - Guang-Liang Wu
- grid.411866.c0000 0000 8848 7685Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 510405 Guangzhou, China ,grid.413402.00000 0004 6068 0570Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, 510120 Guangzhou, China
| | - Li-Jun Qiao
- grid.411866.c0000 0000 8848 7685Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 510405 Guangzhou, China ,grid.413402.00000 0004 6068 0570Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, 510120 Guangzhou, China
| | - Ye-Feng Cai
- grid.411866.c0000 0000 8848 7685Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 510405 Guangzhou, China ,grid.413402.00000 0004 6068 0570Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, 510120 Guangzhou, China
| | - Shi-Jie Zhang
- grid.411866.c0000 0000 8848 7685Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 510405 Guangzhou, China ,grid.413402.00000 0004 6068 0570Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, 510120 Guangzhou, China
| |
Collapse
|
12
|
Madrigal-Bujaidar E, Paniagua-Pérez R, Rendón-Barrón MJ, Morales-González JA, Madrigal-Santillán EO, Álvarez-González I. Investigation of the DNA Damage and Oxidative Effect Induced by Venlafaxine in Mouse Brain and Liver Cells. TOXICS 2022; 10:737. [PMID: 36548570 PMCID: PMC9783611 DOI: 10.3390/toxics10120737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/22/2022] [Accepted: 11/24/2022] [Indexed: 06/17/2023]
Abstract
Venlafaxine is an antidepressant used worldwide. Therefore, studies to confirm its safe use are mandatory. This report evaluated the drug DNA damage capacity in the brain and liver of ICR mice, and its oxidative effect on DNA, lipids, and proteins, as well as the amount of nitrites, also in the brain and liver. Determinations were made at 2, 6, 12, and 24 h post-treatment, excluding DNA oxidation that was observed at 2 h. The tested doses of venlafaxine were 5, 50, and 250 mg/kg. The results showed DNA damage in the brain with the two more elevated doses of venlafaxine at 2 and 6 h post-treatment and also at 12 h in the liver. The comet assay plus the FPG enzyme showed DNA damage in both organs with all doses. The two high doses increased lipoperoxidation in the two tissues from 6 to 12 h post-administration. Protein oxidation increased with the three doses, mainly from 2 to 12 h, and nitrite content was elevated only with the high dose in the liver. The drug was found to affect both tissues, although it was more pronounced in the liver. Interestingly, DNA oxidative damage was observed even with a dose that corresponds to the therapeutic range. The clinical relevance of these findings awaits further investigations.
Collapse
Affiliation(s)
- Eduardo Madrigal-Bujaidar
- Laboratorio de Genética, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Av. Wilfrido Massieu s/n, Zacatenco, Gustavo A. Madero, Ciudad de México 07738, Mexico
| | - Rogelio Paniagua-Pérez
- Instituto Nacional de Rehabilitación, Servicio de Bioquímica. Av., México-Xochimilco 289, Ciudad de México 14389, Mexico
| | - Michael Joshue Rendón-Barrón
- Laboratorio de Genética, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Av. Wilfrido Massieu s/n, Zacatenco, Gustavo A. Madero, Ciudad de México 07738, Mexico
| | - José Antonio Morales-González
- Laboratorio de Medicina de la Conservación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Casco de Santo Tomás, Ciudad de México 11340, Mexico
| | - Eduardo O. Madrigal-Santillán
- Laboratorio de Medicina de la Conservación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Casco de Santo Tomás, Ciudad de México 11340, Mexico
| | - Isela Álvarez-González
- Laboratorio de Genética, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Av. Wilfrido Massieu s/n, Zacatenco, Gustavo A. Madero, Ciudad de México 07738, Mexico
| |
Collapse
|
13
|
Geilen J, Kainz M, Zapletal B, Geleff S, Wisser W, Bohle B, Schweiger T, Schultz MJ, Tschernko E. Unilateral acute lung injury in pig: a promising animal model. J Transl Med 2022; 20:548. [PMID: 36435803 PMCID: PMC9701381 DOI: 10.1186/s12967-022-03753-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 11/04/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Acute lung injury (ALI) occurs in 23% unilateral. Models of unilateral ALI were developed and used previously without clearly demonstrating the strictly unilateral nature and severity of lung injury by the key parameters characterizing ALI as defined by the American Thoracic Society (ATS). Thus, the use of unilateral ALI remained rare despite the innovative approach. Therefore, we developed a unilateral model of ALI and focused on the crucial parameters characterizing ALI. This model can serve for direct comparisons between the injured and intact lungs within single animals, thus, reducing the number of animals required for valid experimental conclusions. METHODS We established the model in nine pigs, followed by an evaluation of key parameters in six pigs (main study). Pigs were ventilated using an adapted left double-lumen tube for lung separation and two ventilators. ALI was induced in the left lung with cyclic rinsing (NaCl 0.9% + Triton® X-100), after which pigs were ventilated for different time spans to test for the timing of ALI onset. Ventilatory and metabolic parameters were evaluated, and bronchoalveolar lavage (BAL) was performed for measurements of inflammatory mediators. Finally, histopathological specimens were collected and examined in respect of characteristics defining the lung injury score (LIS) as suggested by the ATS. RESULTS After adjustments of the model (n = 9) we were able to induce strictly left unilateral ALI in all six pigs of the evaluation study. The median lung injury score was 0.72 (IQR 0.62-0.79) in the left lung vs 0.14 (IQR 0.14-0.16; p < 0.05) in the right lung, confirming unilateral ALI. A significant and sustained drop in pulmonary compliance (Cdyn) of the left lung occurred immediately, whereas Cdyn of the right lung remained unchanged (p < 0.05). BAL fluid concentrations of interleukin-6 and -8 were increased in both lungs. CONCLUSIONS We established a model of unilateral ALI in pigs, confirmed by histopathology, and typical changes in respiratory mechanics and an inflammatory response. This thoroughly evaluated model could serve as a basis for future studies and for comparing pathophysiological and pharmacological changes in the uninjured and injured lung within the same animal.
Collapse
Affiliation(s)
- Johannes Geilen
- Division of Cardiothoracic and Vascular Anesthesia & Critical Care Medicine, Department of Anesthesia, General Intensive Care and Pain Management, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Matthias Kainz
- Division of Cardiothoracic and Vascular Anesthesia & Critical Care Medicine, Department of Anesthesia, General Intensive Care and Pain Management, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Bernhard Zapletal
- Division of Cardiothoracic and Vascular Anesthesia & Critical Care Medicine, Department of Anesthesia, General Intensive Care and Pain Management, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Silvana Geleff
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Wilfried Wisser
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | - Barbara Bohle
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Thomas Schweiger
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Marcus J. Schultz
- Department of Intensive Care, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Edda Tschernko
- Division of Cardiothoracic and Vascular Anesthesia & Critical Care Medicine, Department of Anesthesia, General Intensive Care and Pain Management, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
14
|
Shaban NZ, Mohammed AS, Abu-Serie MM, Maher AM, Habashy NH. Inhibition of oxidative stress, IL-13, and WNT/β-catenin in ovalbumin-sensitized rats by a novel organogel of Punica granatum seed oil saponifiable fraction. Biomed Pharmacother 2022; 154:113667. [PMID: 36942603 DOI: 10.1016/j.biopha.2022.113667] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/31/2022] [Accepted: 09/05/2022] [Indexed: 12/19/2022] Open
Abstract
Bronchial asthma is a chronic inflammatory disease marked by inflammation, oxidative stress, and structural remodeling. Here, we prepared two pomegranate fractions from the seed oil, saponifiable (Sap) and unsaponifiable (UnSap). Two organogels (Orgs) were also formulated with the Sap (Org1) or the UnSap (Org2) fraction and beeswax (BW). All preparations were evaluated in vitro for their antioxidant and anti-inflammatory impacts. The transdermal delivery of the most efficient one was evaluated against ovalbumin (OV)-induced bronchial asthma in rats compared to dexamethasone (DEX). The results showed that the prepared pomegranate fractions and BW had considerable amounts of phenolics (flavonoids and tannins) and triterpenoids. Org1 was shown to be the most effective antioxidant and anti-inflammatory fraction with synergistic activities (combination index, 1), as well as having protective and therapeutic influences on OV-sensitized rats. Org1 inhibited the multiple OV-induced signaling pathways, comprising ROS, WNT/β-catenin, and AKT, with an efficiency superior to DEX. Subsequently, the pro-inflammatory (COX-2, NO, and IL-13), and pro-fibrotic (COL1A1) mediators, oxidative stress, and mucin secretion, were all down-regulated. These outcomes were verified by the histopathological results of lung tissue. Collectively, these outcomes suggest that the transdermal delivery of Org1 to OV-sensitized rats shows promise in the protection and treatment of the pathological hallmarks of asthma.
Collapse
Affiliation(s)
- Nadia Z Shaban
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Alaa S Mohammed
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Marwa M Abu-Serie
- Department of Medical Biotechnology, Genetic Engineering, and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg EL-Arab 21934, Alexandria, Egypt
| | - Adham M Maher
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Noha H Habashy
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt.
| |
Collapse
|
15
|
Ribeiro O, Félix L, Ribeiro C, Castro B, Tiritan ME, Monteiro SM, Carrola JS. Enantioselective Ecotoxicity of Venlafaxine in Aquatic Organisms: Daphnia and Zebrafish. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2022; 41:1851-1864. [PMID: 35452529 DOI: 10.1002/etc.5338] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/16/2022] [Accepted: 03/31/2022] [Indexed: 06/14/2023]
Abstract
Venlafaxine is a chiral antidepressant detected in aquatic compartments. It was recently included in the 3rd Watch List from the European Union. The present study aimed to investigate venlafaxine toxicity effects, targeting possible enantioselective effects, using two aquatic organisms, daphnia (Daphnia magna) and zebrafish (Danio rerio). Specimens were exposed to both racemate, (R,S)-venlafaxine (VEN), and to pure enantiomers. Acute assays with daphnia showed that up to 50 000 μg/L of the (R,S)-VEN induced no toxicity. Organisms were also exposed to sublethal concentrations (25-400 μg/L) of (R,S)-, (R)- and (S)-VEN, for 21 days. No significant effects on mortality, age at first reproduction, and size of the first clutch were observed. However, a decrease in fecundity was observed for both enantiomers at the highest concentration. Regarding zebrafish, the effects of venlafaxine on mortality, embryo development, behavior, biochemistry, and melanin pigmentation were investigated after 96 h of exposure to the range of 0.3-3000 μg/L. (R)-VEN significantly increased the percentage of malformations in comparison with (S)-VEN. Behavior was also enantiomer dependent, with a decrease in the total distance moved and an increase in avoidance behavior observed in organisms exposed to (R)-VEN. Despite the biochemical variations, no changes in redox homeostasis were observed. (R)-VEN also led to an increase in zebrafish pigmentation. The different susceptibility to venlafaxine and enantioselective effects were observed in zebrafish. Our results suggest that at environmental levels (R,S)-VEN and pure enantiomers are not expected to induce harmful effects in both organisms, but (R)-VEN increased malformations in zebrafish larvae, even at reported environmental levels. These results highlight the importance of including enantioselective studies for an accurate risk assessment of chiral pollutants. Environ Toxicol Chem 2022;41:1851-1864. © 2022 SETAC.
Collapse
Affiliation(s)
- Ondina Ribeiro
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes and Alto Douro, Quinta de Prados, Vila Real, Portugal
| | - Luís Félix
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes and Alto Douro, Quinta de Prados, Vila Real, Portugal
- Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production, Vila Real, Portugal
- Institute for Research and Innovation in Health, Laboratory Animal Science, Institute of Molecular and Cell Biology, University of Porto, Porto, Portugal
| | - Cláudia Ribeiro
- Interdisciplinary Center of Marine and Environmental Research, University of Porto, Edifício do Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, Matosinhos, Portugal
- Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, Gandra, Portugal
| | - Bruno Castro
- Department of Biology, Centre of Molecular and Environmental Biology, University of Minho, Braga, Portugal
- Institute of Science and Innovation for Bio-Sustainability, University of Minho, Braga, Portugal
| | - Maria Elizabeth Tiritan
- Interdisciplinary Center of Marine and Environmental Research, University of Porto, Edifício do Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, Matosinhos, Portugal
- Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, Gandra, Portugal
- Organic and Pharmaceutical Chemistry Laboratory, Chemical Sciences Department, Faculty of Pharmacy, University of Porto, Rua Jorge de Viterbo Ferreira, Porto, Portugal
| | - Sandra Mariza Monteiro
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes and Alto Douro, Quinta de Prados, Vila Real, Portugal
- Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production, Vila Real, Portugal
| | - João Soares Carrola
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes and Alto Douro, Quinta de Prados, Vila Real, Portugal
- Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production, Vila Real, Portugal
| |
Collapse
|
16
|
Tao L, Qing Y, Cui Y, Shi D, Liu W, Chen L, Cao Y, Dai Z, Ge X, Zhang L. Lysosomal membrane permeabilization mediated apoptosis involve in perphenazine-induced hepatotoxicity in vitro and in vivo. Toxicol Lett 2022; 367:76-87. [PMID: 35914675 DOI: 10.1016/j.toxlet.2022.07.814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/24/2022] [Accepted: 07/28/2022] [Indexed: 12/11/2022]
Abstract
Antipsychotic drugs represent a class of lysosomotropic drugs widely used in clinical practice. However, the hepatotoxicity of these drugs has been reported in recent years. Therefore, understanding the changes in cellular homeostasis mediated by these drugs is of great significance for revealing the true mechanisms underlying hepatotoxicity. Perphenazine is a classical antipsychotic drug that can reportedly induce extrapyramidal and sympatholytic side effects. The present research focuses on the toxicity effect of perphenazine on normal human hepatocytes. To assess the hepatotoxicity of continuous administration of perphenazine and investigate potential mechanisms related to apoptosis, human normal L02 hepatocytes were exposed to 10-40μM perphenazine in vitro. The results showed that perphenazine inhibited cell viability in a concentration and time-dependent manner. Furthermore, 30μM perphenazine induced intense lysosome vacuolation, impaired lysosomal membrane, and induced lysosomal membrane permeabilization (LMP), ultimately triggering lysosomal cell death in L02 cells. Knockdown cathepsin D(CTSD) also ameliorated perphenazine-induced liver injury via the inhibition of LMP. In vivo, ICR mice received intragastric administration of 10-180mg/kg B.W. perphenazine every other day for 21 days. 180mg/kg perphenazine significantly increased histological injury and aminotransferases compared with control. Taken together, our findings suggest that perphenazine can trigger hepatotoxicity through lysosome disruption both in vitro and in vivo.
Collapse
Affiliation(s)
- Lei Tao
- Nanjing Institute for Food and Drug Control, Jiangsu, Nanjing 211198, China.
| | - Yingjie Qing
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangsu, Nanjing 211198, China.
| | - Yingyue Cui
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangsu, Nanjing 211198, China.
| | - Da Shi
- Nanjing Institute for Food and Drug Control, Jiangsu, Nanjing 211198, China.
| | - Wenting Liu
- Nanjing Institute for Food and Drug Control, Jiangsu, Nanjing 211198, China.
| | - Lei Chen
- Nanjing Institute for Food and Drug Control, Jiangsu, Nanjing 211198, China.
| | - Yu Cao
- Nanjing Institute for Food and Drug Control, Jiangsu, Nanjing 211198, China.
| | - Zhen Dai
- Nanjing Institute for Food and Drug Control, Jiangsu, Nanjing 211198, China.
| | - Xiaoming Ge
- Nanjing Institute for Food and Drug Control, Jiangsu, Nanjing 211198, China.
| | - Ling Zhang
- Nanjing Institute for Food and Drug Control, Jiangsu, Nanjing 211198, China.
| |
Collapse
|
17
|
Asgharzadeh F, Mostafapour A, Ebrahimi S, Amerizadeh F, Sabbaghzadeh R, Hassanian SM, Fakhraei M, Farshbaf A, Ferns GA, Giovannetti E, Avan A, Khazaei M. Inhibition of angiotensin pathway via valsartan reduces tumor growth in models of colorectal cancer. Toxicol Appl Pharmacol 2022; 440:115951. [PMID: 35235860 DOI: 10.1016/j.taap.2022.115951] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 02/09/2022] [Accepted: 02/23/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND Overexpression of the angiotensin-II receptor and renin-angiotensin system (RAS) has been reported in several malignancies, including colorectal-cancer (CRC), indicating its potential value as a therapeutic target. Here we explored the impact of targeting the RAS using an angiotensin II receptor blocker, valsartan, alone and its combination with Fluorouracil (5-FU) in in vitro and in vivo models of CRC. METHODS Anti-proliferative activity of valsartan was evaluated in 2-/3-dimensional in vitro and in vivo CRC mouse models. The anti-migratory effects of this agent was assessed by wound-healing assay, while apoptosis was studied using 4',6-diamidino-2-phenylindole or DAPI staining, and staining with Annexin-V-fluorescein isothiocyanate with analysis using FACS. Gene-expression was determined at mRNA and protein levels. We further evaluated the anti-inflammatory properties of valsartan by histological analysis and the measurement of oxidative/antioxidant markers. Gelatin zymography was used to measure matrix metalloproteinase-2 and -9 activity (MMP-2 and 9). RESULTS Valsartan suppressed CRC cell-growth and synergistically enhanced the anti-tumor-activities of 5-FU by induction of apoptosis, BAX, BCL2, P53 and modulation of the cell cycle. Valsartan inhibited the cell migration by perturbation of MMP2/9. Furthermore, valsartan inhibited tumor-growth, and this was more pronounced when using the valsartan/5-FU combination. The plausible mechanism for this is via the induction of ROS and down-regulation of SOD, thiol/catalase as well as VEGF. Valsartan may protect cells against intestinal fibrosis by modulation of pro-fibrotic and pro-inflammatory factors including interleukins and Col1A1 expression. CONCLUSIONS Our findings demonstrated that targeting RAS pathway using Valsartan interferes with cell-proliferation, induces apoptosis, reduces migration and synergistically interacts with 5-FU, supporting further studies on this new therapeutic approach for colorectal cancer.
Collapse
Affiliation(s)
- Fereshteh Asgharzadeh
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Asma Mostafapour
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Safieh Ebrahimi
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Forouzan Amerizadeh
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Basic Medical Sciences Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reihaneh Sabbaghzadeh
- Department of Biology, Faculty of Science, Hakim Sabzevari University, Sabzevar 96179-76487, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Fakhraei
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alieh Farshbaf
- Dental Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Basic Medical Sciences Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetics Research center, Mashhad University of Medical Sciences, Mashhad, Iran..
| | - Majid Khazaei
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
18
|
Fang L, Wang S, Cao L, Yao K. Early intervention of acute liver injury related to venlafaxine: A case report. Medicine (Baltimore) 2021; 100:e28140. [PMID: 34889278 PMCID: PMC8663904 DOI: 10.1097/md.0000000000028140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 11/17/2021] [Indexed: 01/05/2023] Open
Abstract
RATIONALE Drug-induced liver injury (DILI) is the leading cause of acute liver injury (ALI), market withdrawal of a drug, and rejection of applications for marketing licenses. The incidence of DILI is very low, with a value between 1 and 19 per 100,000 patient years. All antidepressants may induce DILI even at low therapeutic doses. In this report, we present a case of ALI after venlafaxine administration. PATIENT CONCERNS A 27-year-old Chinese Han woman was admitted for depression. Several serum liver function indices in this patient were abnormal after antidepressant treatment. The Roussel Uclaf Causality Assessment Method (RUCAM) causality assessment score was 8, and the R value was 31.18. DIAGNOSES The patient was diagnosed with hepatocellular ALI, which was derived from venlafaxine-related adverse events. INTERVENTIONS First, all medications were stopped to block the progression of DILI. Then, a hepatoprotective strategy and proper psychological treatment were performed to recover the impaired hepatic function. OUTCOMES Liver function was fully recovered as indicated by liver function indices and ultrasound imaging. LESSONS The possibility of DILI should not be overlooked during the long-term use of antipsychotic drugs. In response, regular liver function monitoring should be performed in a timely manner to avoid missing diagnoses and delayed treatment. Furthermore, the necessary medical treatment needs to be conducted after the occurrence of ALI.
Collapse
Affiliation(s)
- Lin Fang
- Department of Clinical Psychology, The Affiliated Wuxi Mental Health Center with Nanjing Medical University, Wuxi, Jiangsu 214151, China
| | - Shushan Wang
- Department of Pharmacy, The Affiliated Wuxi Mental Health Center with Nanjing Medical University, Wuxi, Jiangsu 214151, China
| | - Leiming Cao
- Department of Clinical Psychology, The Affiliated Wuxi Mental Health Center with Nanjing Medical University, Wuxi, Jiangsu 214151, China
| | - Kun Yao
- Department of Clinical Psychology, The Affiliated Wuxi Mental Health Center with Nanjing Medical University, Wuxi, Jiangsu 214151, China
| |
Collapse
|
19
|
Li Y, Wang Y, Zhang N, Zhou S, Peng Y, Zheng J. Glutathione conjugation and protein modification resulting from metabolic activation of venlafaxine in vitro and in vivo. Xenobiotica 2021; 51:1303-1317. [PMID: 34160341 DOI: 10.1080/00498254.2021.1946204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Venlafaxine (VLF), an antidepressant agent, is widely used to combat major depressive disorders, particularly for the treatment of selective serotonin reuptake inhibitor-resistant depression. VLF has been shown to cause liver injury. The present study aimed to investigate the metabolic activation of VLF and explore the mechanisms of hepatotoxicity induced by VLF.One glutathione (GSH) conjugate and one cysteine conjugate were both detected in mouse and human liver microsomal incubations containing VLF and GSH or cysteine. The two conjugates were also detected in cultured mouse primary hepatocytes and bile of rats after exposure to VLF. The in vitro and in vivo studies demonstrated that VLF was metabolized to a quinone methide intermediate reactive to GSH and cysteine residues of hepatic protein. The observed protein covalent binding revealed dose-dependency. The metabolic activation of VLF was P450-dependent, and CYP3A4 was found as the predominant enzyme involved in the bioactivation process.These findings facilitate better understanding of the metabolic activation-hepatotoxicity relationship of VLF and provide chemists with information about new potential structural alerts during drug design process.
Collapse
Affiliation(s)
- Yilin Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Yang Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Na Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Shenzhi Zhou
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Ying Peng
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Jiang Zheng
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China.,State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou 550025, P. R. China.,Key Laboratory of Environmental Pollution, Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, P. R. China
| |
Collapse
|
20
|
Ommati MM, Niknahad H, Farshad O, Azarpira N, Heidari R. In Vitro and In Vivo Evidence on the Role of Mitochondrial Impairment as a Mechanism of Lithium-Induced Nephrotoxicity. Biol Trace Elem Res 2021; 199:1908-1918. [PMID: 32712907 DOI: 10.1007/s12011-020-02302-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/21/2020] [Indexed: 02/08/2023]
Abstract
Lithium is abundantly administered against bipolar disorder. On the other hand, the lithium-induced renal injury is a clinical complication which commonly reveals as drug-induced diabetes insipidus. However, lithium-induced cytotoxicity might also play a role in the adverse effects of this drug on the kidney. There is no clear cellular and molecular mechanism(s) for lithium-induced nephrotoxicity. The current study was designed to assess the effect of lithium on kidney tissue oxidative stress biomarkers and mitochondrial function and its relevance to drug-induced nephrotoxicity and electrolyte imbalance. Rats were treated with lithium (lithium carbonate, 25 and 50 mg/kg/day, i.p., for 28 consecutive days). Kidney mitochondria were also isolated from rats and exposed to increasing concentrations of lithium (0.01-10 mM). Serum and urine biomarkers of kidney injury, kidney tissue markers of oxidative stress, and renal histopathological changes were assessed. Moreover, several mitochondrial indices were monitored. Lithium-induced renal injury revealed a significant increase in urine and serum biomarkers of renal impairment. Lithium caused an increase in the kidney reactive oxygen species (ROS) level and lipid peroxidation (LPO). Renal glutathione (GSH) reservoirs were also depleted, and tissue antioxidant capacity decreased in lithium-treated animals. Significant tissue histopathological changes, including necrosis, Bowman capsule dilation, and interstitial inflammation, were evident in lithium-treated animals. On the other hand, significant alterations in kidney mitochondrial function were detected in lithium-treated groups. These data mention oxidative stress, mitochondrial dysfunction, and cellular energy crisis as the potential primary mechanisms for lithium-induced renal injury.
Collapse
Affiliation(s)
- Mohammad Mehdi Ommati
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Hossein Niknahad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, P. O. Box 1583; 71345. Roknabad, Karafarin St., Shiraz, Fars, Iran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Omid Farshad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, P. O. Box 1583; 71345. Roknabad, Karafarin St., Shiraz, Fars, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, P. O. Box 1583; 71345. Roknabad, Karafarin St., Shiraz, Fars, Iran.
| |
Collapse
|
21
|
Kaur S, Kaur A, Jaswal N, Aniqa A, Sadwal S, Bharati S. Selenium attenuates venlafaxine hydrochloride-induced testicular damage in mice via modulating oxidative stress and apoptosis. Andrologia 2021; 53:e14050. [PMID: 33733493 DOI: 10.1111/and.14050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/17/2021] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
The present study assessed the effect of selenium (Se) supplementation on Venlafaxine hydrochloride (VH)-induced testicular toxicity. Mice were segregated into Group I (C), Group II (0.5 ppm Se), Group III (VH at a dose 60 mg/kg b.w.) and Group IV (Se was given as per Group II, and VH was given as per Group III). After 10 weeks, sperm parameters, histology, sperm cell counts, antioxidants activities, apoptotic proteins and molecular analysis of testicular tissue were evaluated. Group III had significantly lower sperm concentration (from 2.17 ± 0.28 to 1.04 ± 0.22) and sperm motility (from 68.04 ± 5.5 to 21.47 ± 5.21), and showed an extensive vacuolisation in the germinal epithelium, abnormal basement membrane, and reduced germ cell number as compared to Group I. However, selenium supplementation in Group IV substantially increased sperm concentration (1.47 ± 0.48) and motility (33.27 ± 8.66), improved the histoarchitecture and repopulated the germ cells as observed by raised numbers of spermatogonia, spermatocytes, round spermatids and elongated spermatids contrasted to Group III. Group IV also showed a noteworthy decreased ROS, LPO levels, as well as expressions of Bax, caspase-9, and caspase-3 and increased the SOD, CAT, GPx, and GSH activities as well the expression of Bcl-2 as compared to Group III. This effect was further supported by FTIR analysis for nucleic acids. Thus, selenium supplementation showed significant protection against VH-induced testicular toxicity.
Collapse
Affiliation(s)
| | - Amarjit Kaur
- Department of Chemistry, Panjab University, Chandigarh, India
| | - Nisha Jaswal
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Aniqa Aniqa
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Shilpa Sadwal
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Sanjay Bharati
- Department of Nuclear Medicine, Manipal College of Health Professions, Manipal Academy of Higher Education, Udupi, India
| |
Collapse
|
22
|
Hashemzehi M, Rahmani F, Khoshakhlagh M, Avan A, Asgharzadeh F, Barneh F, Moradi-Marjaneh R, Soleimani A, Fiuji H, Ferns GA, Ryzhikov M, Jafari M, Khazaei M, Hassanian SM. Angiotensin receptor blocker Losartan inhibits tumor growth of colorectal cancer. EXCLI JOURNAL 2021; 20:506-521. [PMID: 33883980 PMCID: PMC8056058 DOI: 10.17179/excli2020-3083] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 02/18/2021] [Indexed: 12/22/2022]
Abstract
The renin-angiotensin system (RAS) is up-regulated in patients with colorectal cancer (CRC) and is reported to be associated with poor prognosis and chemo-resistance. Here we explored the therapeutic potential of targeting RAS in CRC using Losartan, an angiotensin receptor blocker. An integrative-systems biology approach was used to explore a proteome-level dataset of a gene signature that is modulated by Losartan. The anti-proliferative activity of Losartan was evaluated using 2- and 3-dimensional cell culture models. A xenograft model of colon cancer was used to investigate tumor growth with Losartan alone and in combination with 5-FU followed by histological staining (Hematoxylin & Eosin and Masson trichrome staining), biochemical analyses, gene expression analyses by RT-PCR, western blot/IHC, or MMP Gelatin Zymography studies. Effects on cell cycle and cell death were assessed by flow cytometry. Losartan inhibited cell growth and suppressed cell cycle progression, causing an increase in CRC cells in the G1 phase. Losartan significantly reduced tumor growth and enhanced tumor cell necrosis. An impact on the inflammatory response, including up-regulation of pro-inflammatory cytokines and chemokines in CRC cells are potential mechanisms that could partially explain Losartan's anti-proliferative effects. Moreover, metastasis and angiogenesis were reduced in Losartan-treated mice as observed by inhibited matrix metalloproteinase-2 and -9 activities and decreased tumor vasculature. These data demonstrate the therapeutic potential of combining chemotherapeutic regimens with Losartan to synergistically enhance its activity and target the renin-angiotensin system as a new approach in colorectal cancer treatment.
Collapse
Affiliation(s)
- Milad Hashemzehi
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Iranshahr University of Medical Sciences, Iranshahr, Iran.,Tropical and Communicable Diseases Research Centre, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Farzad Rahmani
- Iranshahr University of Medical Sciences, Iranshahr, Iran.,Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahdieh Khoshakhlagh
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fereshteh Asgharzadeh
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farnaz Barneh
- Faculty of Paramedical Sciences, Beheshti University of Medical Sciences, Tehran, Iran; Current address: Princess Maxima Center for Pediatric Oncology, 3584, CS, Utrecht, The Netherlands
| | - Reyhaneh Moradi-Marjaneh
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Atena Soleimani
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Fiuji
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | | | - Mohieddin Jafari
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science, University of Helsinki, Finland
| | - Majid Khazaei
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
23
|
Todorović Vukotić N, Đorđević J, Pejić S, Đorđević N, Pajović SB. Antidepressants- and antipsychotics-induced hepatotoxicity. Arch Toxicol 2021; 95:767-789. [PMID: 33398419 PMCID: PMC7781826 DOI: 10.1007/s00204-020-02963-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023]
Abstract
Drug-induced liver injury (DILI) is a serious health burden. It has diverse clinical presentations that can escalate to acute liver failure. The worldwide increase in the use of psychotropic drugs, their long-term use on a daily basis, common comorbidities of psychiatric and metabolic disorders, and polypharmacy in psychiatric patients increase the incidence of psychotropics-induced DILI. During the last 2 decades, hepatotoxicity of various antidepressants (ADs) and antipsychotics (APs) received much attention. Comprehensive review and discussion of accumulated literature data concerning this issue are performed in this study, as hepatotoxic effects of most commonly prescribed ADs and APs are classified, described, and discussed. The review focuses on ADs and APs characterized by the risk of causing liver damage and highlights the ones found to cause life-threatening or severe DILI cases. In parallel, an overview of hepatic oxidative stress, inflammation, and steatosis underlying DILI is provided, followed by extensive review and discussion of the pathophysiology of AD- and AP-induced DILI revealed in case reports, and animal and in vitro studies. The consequences of some ADs and APs ability to affect drug-metabolizing enzymes and therefore provoke drug–drug interactions are also addressed. Continuous collecting of data on drugs, mechanisms, and risk factors for DILI, as well as critical data reviewing, is crucial for easier DILI diagnosis and more efficient risk assessment of AD- and AP-induced DILI. Higher awareness of ADs and APs hepatotoxicity is the prerequisite for their safe use and optimal dosing.
Collapse
Affiliation(s)
- Nevena Todorović Vukotić
- Department of Molecular Biology and Endocrinology, "Vinča" Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 12-14 Mike Petrovića Alasa, P.O. Box 522-090, 11000, Belgrade, Serbia.
| | - Jelena Đorđević
- Institute of Physiology and Biochemistry "Ivan Đaja", Faculty of Biology, University of Belgrade, 16 Studentski Trg, 11000, Belgrade, Serbia
| | - Snežana Pejić
- Department of Molecular Biology and Endocrinology, "Vinča" Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 12-14 Mike Petrovića Alasa, P.O. Box 522-090, 11000, Belgrade, Serbia
| | - Neda Đorđević
- Department of Molecular Biology and Endocrinology, "Vinča" Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 12-14 Mike Petrovića Alasa, P.O. Box 522-090, 11000, Belgrade, Serbia
| | - Snežana B Pajović
- Department of Molecular Biology and Endocrinology, "Vinča" Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 12-14 Mike Petrovića Alasa, P.O. Box 522-090, 11000, Belgrade, Serbia.,Faculty of Medicine, University of Niš, 81 Blvd. Dr. Zorana Đinđića, 18000, Niš, Serbia
| |
Collapse
|
24
|
Allen J, Caruncho HJ, Kalynchuk LE. Severe life stress, mitochondrial dysfunction, and depressive behavior: A pathophysiological and therapeutic perspective. Mitochondrion 2020; 56:111-117. [PMID: 33220501 DOI: 10.1016/j.mito.2020.11.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 10/28/2020] [Accepted: 11/11/2020] [Indexed: 01/11/2023]
Abstract
Mitochondria are responsible for providing our cells with energy, as well as regulating oxidative stress and apoptosis, and considerable evidence demonstrates that mitochondria-related alterations are prevalent during chronic stress and depression. Here, we discuss how chronic stress may induce depressive behavior by potentiating mitochondrial allostatic load, which ultimately decreases energy production, elevates the generation of harmful reactive oxygen species, damages mitochondrial DNA and increases membrane permeability and pro-apoptotic factor release. We also discuss how mitochondrial insults can exacerbate the immune response, contributing to depressive symptomology. Furthermore, we illustrate how depression symptoms are associated with specific mitochondrial defects, and how targeting of these defects with pharmacological agents may be a promising avenue for the development of novel, more efficacious antidepressants. In summary, this review supports the notion that severe psychosocial stress induces mitochondrial dysfunction, thereby increasing the vulnerability to developing depressive symptoms.
Collapse
Affiliation(s)
- Josh Allen
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
| | - Hector J Caruncho
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Lisa E Kalynchuk
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| |
Collapse
|
25
|
Mousavi K, Niknahad H, Ghalamfarsa A, Mohammadi H, Azarpira N, Ommati MM, Heidari R. Taurine mitigates cirrhosis-associated heart injury through mitochondrial-dependent and antioxidative mechanisms. Clin Exp Hepatol 2020; 6:207-219. [PMID: 33145427 PMCID: PMC7592093 DOI: 10.5114/ceh.2020.99513] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 04/09/2020] [Indexed: 12/15/2022] Open
Abstract
Cirrhosis-induced heart injury and cardiomyopathy is a serious consequence of this disease. It has been shown that bile duct ligated (BDL) animals could serve as an appropriate experimental model to investigate heart tissue injury in cirrhosis. The accumulation of cytotoxic chemicals (e.g., bile acids) could also adversely affect the heart tissue. Oxidative stress and mitochondrial impairment are the most prominent mechanisms of bile acid cytotoxicity. Taurine (Tau) is the most abundant non-protein amino acid in the human body. The cardioprotective effects of this amino acid have repeatedly been investigated. In the current study, it was examined whether mitochondrial dysfunction and oxidative stress are involved in the pathogenesis of cirrhosis-induced heart injury. Rats underwent BDL surgery. BDL animals received Tau (50, 100, and 500 mg/kg, i.p.) for 42 consecutive days. A significant increase in oxidative stress biomarkers was detected in the heart tissue of BDL animals. Moreover, it was found that heart tissue mitochondrial indices of functionality were deteriorated in the BDL group. Tau treatment significantly decreased oxidative stress and improved mitochondrial function in the heart tissue of cirrhotic animals. These data provide clues for the involvement of mitochondrial impairment and oxidative stress in the pathogenesis of heart injury in BDL rats. On the other hand, Tau supplementation could serve as an effective ancillary treatment against BDL-associated heart injury. Mitochondrial regulating and antioxidative properties of Tau might play a fundamental role in its mechanism of protective effects in the heart tissue of BDL animals.
Collapse
Affiliation(s)
- Khadijeh Mousavi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Niknahad
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amin Ghalamfarsa
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamidreza Mohammadi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
26
|
Mehdi H, Bragg LM, Servos MR, Craig PM. Multiple Stressors in the Environment: The Effects of Exposure to an Antidepressant (Venlafaxine) and Increased Temperature on Zebrafish Metabolism. Front Physiol 2019; 10:1431. [PMID: 31803073 PMCID: PMC6877669 DOI: 10.3389/fphys.2019.01431] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/04/2019] [Indexed: 12/18/2022] Open
Abstract
Aquatic organisms are continuously exposed to multiple environmental stressors working cumulatively to alter ecosystems. Wastewater-dominated environments are often riddled by a myriad of stressors, such as chemical and thermal stressors. The objective of this study was to examine the effects of an environmentally relevant concentration of a commonly prescribed antidepressant, venlafaxine (VFX) [1.0 μg/L], in addition to a 5°C increase in water temperature on zebrafish metabolism. Fish were chronically exposed (21 days) to one of four conditions: (i) 0 μg/L VFX at 27°C; (ii) 1.0 μg/L VFX at 27°C; (iii) 0 μg/L VFX at 32°C; (iv) 1.0 μg/L VFX at 32°C. Following exposure, whole-body metabolism was assessed by routine metabolic rate (RMR) measurements, whereas tissue-specific metabolism was assessed by measuring the activities of major metabolic enzymes in addition to glucose levels in muscle. RMR was significantly higher in the multi-stressed group relative to Control. The combination of both stressors resulted in elevated pyruvate kinase activity and glucose levels, while lipid metabolism was depressed, as measured by 3-hydroxyacyl CoA dehydrogenase activity. Citrate synthase activity increased with the onset of temperature, but only in the group treatment without VFX. Catalase activity was also elevated with the onset of the temperature stressor, however, that was not the case for the multi-stressed group, potentially indicating a deleterious effect of VFX on the anti-oxidant defense mechanism. The results of this study highlight the importance of multiple-stressor research, as it able to further bridge the gap between field and laboratory studies, as well as have the potential of yielding surprising results that may have not been predicted using a conventional single-stressor approach.
Collapse
Affiliation(s)
- Hossein Mehdi
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | | | | | | |
Collapse
|
27
|
Ommati MM, Farshad O, Jamshidzadeh A, Heidari R. Taurine enhances skeletal muscle mitochondrial function in a rat model of resistance training. PHARMANUTRITION 2019. [DOI: 10.1016/j.phanu.2019.100161] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
28
|
Emadi E, Abdoli N, Ghanbarinejad V, Mohammadi HR, Mousavi Mobarakeh K, Azarpira N, Mahboubi Z, Niknahad H, Heidari R. The potential role of mitochondrial impairment in the pathogenesis of imatinib-induced renal injury. Heliyon 2019; 5:e01996. [PMID: 31294126 PMCID: PMC6595238 DOI: 10.1016/j.heliyon.2019.e01996] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 06/10/2019] [Accepted: 06/19/2019] [Indexed: 12/21/2022] Open
Abstract
Imatinib is a tyrosine kinase inhibitor widely administered against chronic myeloid leukemia. On the other hand, drug-induced kidney proximal tubular injury, electrolytes disturbances, and renal failure is a clinical complication associated with imatinib therapy. There is no precise cellular mechanism(s) for imatinib-induced renal injury. The current investigation aimed to evaluate the role of mitochondrial dysfunction and oxidative stress in the pathogenesis of imatinib nephrotoxicity. Rats received imatinib (50 and 100 mg/kg, oral, 14 consecutive days). Serum and urine biomarkers of renal injury and markers of oxidative stress in the kidney tissue were assessed. Moreover, kidney mitochondria were isolated, and mitochondrial indices, including mitochondrial depolarization, dehydrogenases activity, mitochondrial permeabilization, lipid peroxidation (LPO), mitochondrial glutathione levels, and ATP content were determined. A significant increase in serum (Creatinine; Cr and blood urea nitrogen; BUN) and urine (Glucose, protein, gamma-glutamyl transferase; γ-GT, and alkaline phosphatase; ALP) biomarkers of renal injury, as well as serum electrolytes disturbances (hypokalemia and hypophosphatemia), were evident in imatinib-treated animals. On the other hand, imatinib (100 mg/kg) caused an increase in kidney ROS and LPO. Renal tubular interstitial nephritis, tissue necrosis, and atrophy were evident as tissue histopathological changes in imatinib-treated rats. Mitochondrial parameters were also adversely affected by imatinib administration. These data represent mitochondrial impairment, renal tissue energy crisis, and oxidative stress as possible mechanisms involved in the pathogenesis of imatinib-induced renal injury and serum electrolytes disturbances.
Collapse
Affiliation(s)
- Ehsan Emadi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Narges Abdoli
- Iran Food and Drug Administration (IFDA), Ministry of Health, Tehran, Iran
| | - Vahid Ghanbarinejad
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamid Reza Mohammadi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Khadijeh Mousavi Mobarakeh
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Mahboubi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Niknahad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
29
|
Ghanbarinejad V, Ahmadi A, Niknahad H, Ommati MM, Heidari R. Carnosine Mitigates Manganese Mitotoxicity in an In Vitro Model of Isolated Brain Mitochondria. Adv Pharm Bull 2019; 9:294-301. [PMID: 31380256 PMCID: PMC6664115 DOI: 10.15171/apb.2019.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 12/30/2018] [Accepted: 04/14/2019] [Indexed: 12/12/2022] Open
Abstract
Purpose: Manganese (Mn) is a neurotoxic chemical which induces a wide range of complications in the brain tissue. Impaired locomotor activity and cognitive dysfunction are associated with high brain Mn content. At the cellular level, mitochondria are potential targets for Mn toxicity. Carnosine is a dipeptide abundantly found in human brain. Several pharmacological properties including mitochondrial protecting and antioxidative effects have been attributed to carnosine. The current study aimed to evaluate the effect of carnosine treatment on Mn-induced mitochondrial dysfunction in isolated brain mitochondria. Methods: Mice brain mitochondria were isolated based on the differential centrifugation method and exposed to increasing concentrations of Mn (10 µM-10 mM). Carnosine (1 mM) was added as the protective agent. Mitochondrial indices including mitochondrial depolarization, reactive oxygen species (ROS) formation, mitochondrial dehydrogenases activity, ATP content, and mitochondrial swelling and permeabilization were assessed. Results: Significant deterioration in mitochondrial indices were evident in Mn-exposed brain mitochondria. On the other hand, it was found that carnosine (1 mM) treatment efficiently prevented Mn-induced mitochondrial impairment. Conclusion: These data propose mitochondrial protection as a fundamental mechanism for the effects of carnosine against Mn toxicity. Hence, this peptide might be applicable against Mn neurotoxicity with different etiologies (e.g., in cirrhotic patients).
Collapse
Affiliation(s)
- Vahid Ghanbarinejad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Asrin Ahmadi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Niknahad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Mehdi Ommati
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
30
|
Ammonia-induced mitochondrial impairment is intensified by manganese co-exposure: relevance to the management of subclinical hepatic encephalopathy and cirrhosis-associated brain injury. Clin Exp Hepatol 2019; 5:109-117. [PMID: 31501786 PMCID: PMC6728860 DOI: 10.5114/ceh.2019.85071] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 10/29/2018] [Indexed: 12/30/2022] Open
Abstract
Aim of the study Hepatic encephalopathy (HE) is a neuropsychiatric syndrome ensuing from liver failure. The liver is the major site of ammonia detoxification in the human body. Hence, acute and chronic liver dysfunction can lead to hyperammonemia. Manganese (Mn) is a trace element incorporated in several physiological processes in the human body. Mn is excreted through bile. It has been found that cirrhosis is associated with hyperammonemia as well as body Mn accumulation. The brain is the primary target organ for both ammonia and Mn toxicity. On the other hand, brain mitochondria impairment is involved in the mechanism of Mn and ammonia neurotoxicity. Material and methods The current study was designed to evaluate the effect of Mn and ammonia and their combination on mitochondrial indices of functionality in isolated brain mitochondria. Isolated brain mitochondria were exposed to increasing concentrations of ammonia and Mn alone and/or in combination and several mitochondrial indices were assessed. Results The collapse of mitochondrial membrane potential, increased mitochondrial permeabilization, reactive oxygen species formation, and a significant decrease in mitochondrial dehydrogenase activity and ATP content were evident in Mn-exposed (0.005-1 mM) brain mitochondria. On the other hand, ammonia (0.005-0.5 mM) caused no significant changes in brain mitochondrial function. It was found that co-exposure of the brain mitochondria to Mn and ammonia causes more evident mitochondrial impairment in comparison with Mn and/or ammonia alone. Conclusions These data indicate additive toxicity of ammonia and Mn in isolated brain mitochondria exposed to these neurotoxins.
Collapse
|
31
|
Heidari R, Arabnezhad MR, Ommati MM, Azarpira N, Ghodsimanesh E, Niknahad H. Boldine Supplementation Regulates Mitochondrial Function and Oxidative Stress in a Rat Model of Hepatotoxicity. PHARMACEUTICAL SCIENCES 2019. [DOI: 10.15171/ps.2019.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Background: The xenobiotics-induced liver injury is a clinical complication. Hence, finding new hepatoprotective strategies has clinical value. Oxidative stress and its subsequent complications are major mechanisms involved in xenobiotics-induced hepatotoxicity. Boldine is one of the most potent antioxidant molecules widely investigated for its protective properties in different experimental models. In the current study, the hepatoprotective properties of boldine and its potential mechanisms of hepatoprotection have been investigated. Methods: Rats received thioacetamide (TAA; 200 mg/kg, i.p) as a model of acute liver injury. Boldine (5, 10, 1nd 20 mg/kg; 24 hours intervals; oral) was administered as the hepatoprotective agent. Results: Liver injury was evident in TAA-treated animals (48 hours after TAA exposure) as a severe increase in serum level of liver injury biomarkers and histopathological alterations. Moreover, markers of oxidative stress were increased in liver tissue of TAA-treated rats. Assessment of mitochondrial indices of functionality revealed a significant decrease in mitochondrial dehydrogenases activity, the collapse of mitochondrial membrane potential, mitochondrial swelling and depletion of ATP content. It was found that boldine supplementation mitigated liver tissue markers of oxidative stress and improved mitochondrial indices of functionality in TAA-treated animals. Conclusion: The hepatoprotective properties of boldine might primarily rely on antioxidant and mitochondria protecting effects of this alkaloid.
Collapse
Affiliation(s)
- Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Reza Arabnezhad
- Pharmacology and Toxicology Department, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Mehdi Ommati
- Pharmacology and Toxicology Department, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elham Ghodsimanesh
- Pharmacology and Toxicology Department, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Niknahad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmacology and Toxicology Department, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
32
|
Heidari R, Behnamrad S, Khodami Z, Ommati MM, Azarpira N, Vazin A. The nephroprotective properties of taurine in colistin-treated mice is mediated through the regulation of mitochondrial function and mitigation of oxidative stress. Biomed Pharmacother 2019; 109:103-111. [DOI: 10.1016/j.biopha.2018.10.093] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/14/2018] [Accepted: 10/15/2018] [Indexed: 12/13/2022] Open
|
33
|
Eftekhari A, Heidari R, Ahmadian E, Eghbal MA. Cytoprotective Properties of Carnosine against Isoniazid-Induced Toxicity in Primary Cultured Rat Hepatocytes. PHARMACEUTICAL SCIENCES 2018. [DOI: 10.15171/ps.2018.38] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Background: Drug-induced liver injury is a critical clinical complication. Hence, finding new and safe protective agents with potential clinical application is of value. Isoniazid (INH) is an antituberculosis agent widely used against Mycobacterium tuberculosis infection in human. On the other hand, hepatotoxicity is a clinical complication associated with isoniazid therapy. Oxidative stress and its associated events are major mechanisms identified for INH-induced liver injury. Carnosine is an endogenously found peptide widely investigated for its hepatoprotective effects. On the other hand, robust antioxidant and cytoprotective effects have been attributed to this peptide. Methods: The current study designed to evaluate the potential cytoprotective properties of carnosine against INH-induced cytotoxicity in drug-exposed primary cultured rat hepatocytes. Primary cultured rat hepatocytes were incubated with INH (1.2 mM). Results: INH treatment caused significant increase in cell death and lactate dehydrogenase (LDH) release. On the other hand, it was found that markers of oxidative stress including reactive oxygen species were significantly increased in INH-treated cells. Cellular glutathione reservoirs were also depleted in INH-treated group. Carnosine treatment (50 and 100 µM) significantly diminished INH-induced oxidative stress and cytotoxicity. Conclusion: These data mention carnosine as a potential protective agent with therapeutic capability against INH hepatotoxicity.
Collapse
Affiliation(s)
- Aziz Eftekhari
- Pharmacology and Toxicology Department, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elham Ahmadian
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Ali Eghbal
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
34
|
Heidari R, Ahmadi A, Mohammadi H, Ommati MM, Azarpira N, Niknahad H. Mitochondrial dysfunction and oxidative stress are involved in the mechanism of methotrexate-induced renal injury and electrolytes imbalance. Biomed Pharmacother 2018; 107:834-840. [DOI: 10.1016/j.biopha.2018.08.050] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/10/2018] [Accepted: 08/10/2018] [Indexed: 12/12/2022] Open
|
35
|
Effect of silver nanoparticles in the induction of apoptosis on human hepatocellular carcinoma (HepG2) cell line. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 93:465-471. [PMID: 30274079 DOI: 10.1016/j.msec.2018.08.027] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 07/14/2018] [Accepted: 08/08/2018] [Indexed: 01/04/2023]
Abstract
Silver nanoparticles (Ag NPs) serve numerous chief functions in cosmetics, engineering, textile, food technology and medicine. These nanoparticles are also utilized in the pharmaceutical industry particularly in the production of novel antimicrobial agents. However, despite the various studies of Ag NPs induced toxicity, there is a lack of information concerning cellular toxicity mechanisms of these nanoparticles on human cells. In the current project, we investigate the anti-cancer effects of Ag NPs in HepG2 (liver hepatocellular adenocarcinoma) cells. The mean particle size and morphology for the prepared nanoparticles were determined by dynamic light scattering (DLS) and transmission electron microscopy (TEM), respectively. Cell viability, reactive oxygen species (ROS) formation, cytochrome c amount and expression level of BAX/CASP 3/CASP 8/CASP 9 were assayed in HepG2 cells after incubation with Ag NPs. The prepared nanoparticles showed the mean particle size of 30.71 nm with polydispersity index (PDI) of 0.21. Our results revealed decreased cell viability in a concentration-dependent manner and the IC50 of 75 μg/mL for Ag NPs. Ag NPs cytotoxicity was associated with induction of ROS and cell apoptosis in HepG2 cell line. According to our findings, Ag NPs could be considered as potential chemotherapeutic agents in the treatment of liver hepatocellular carcinoma.
Collapse
|
36
|
Eftekhari A, Dizaj SM, Chodari L, Sunar S, Hasanzadeh A, Ahmadian E, Hasanzadeh M. The promising future of nano-antioxidant therapy against environmental pollutants induced-toxicities. Biomed Pharmacother 2018; 103:1018-1027. [DOI: 10.1016/j.biopha.2018.04.126] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 04/13/2018] [Accepted: 04/17/2018] [Indexed: 12/24/2022] Open
|
37
|
Yazdani J, Ahmadian E, Sharifi S, Shahi S, Maleki Dizaj S. A short view on nanohydroxyapatite as coating of dental implants. Biomed Pharmacother 2018; 105:553-557. [PMID: 29886376 DOI: 10.1016/j.biopha.2018.06.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 06/01/2018] [Accepted: 06/02/2018] [Indexed: 02/01/2023] Open
Abstract
INTRODUCTION Titanium based (Ti-based) materials have been used as dental implants due to their high biocompatibility, good mechanical strength and ideal osseointegration properties. Osseointegration of an implant is dependent on surface characteristics such as surface chemistry and topography. Nanotechnology has presented new and interesting applications in dentistry in recent years. The presence of nanoparticles on the implant surface can affect both the topography and surface chemistry, leading to different and outstanding specifications for implant. METHOD A literature review was performed in electronic databases by means of MeSH keywords to collect relevant published literature in English about the effect of nanohydroxyapatite on osseointegration of titanium implants. No limitations on publication date were imposed. Data regarding titanium implants; nanotechnology; nanohydroxyapatite; osseointegration and cell attachment were collected and reviewed. RESULTS AND CONCLUSION According to reviewed literature, nanohydroxyapatites have a nanostructured surface with higher surface area and then higher reactivity, letting them to bind to bone creating a biomimetic coating on implants. However, more studies are needed on the cell-substrate interface to develop an effective implant due to the interaction of the cells and the biomaterial surface after the implantation.
Collapse
Affiliation(s)
- Javad Yazdani
- Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Ahmadian
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Simin Sharifi
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahriar Shahi
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Faculty of Dentistry, Department of Endodontics, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Solmaz Maleki Dizaj
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
38
|
Paulis MG, Hafez EM, El-Tahawy NF, Aly MKM. Toxicological Assessment of Venlafaxine: Acute and Subchronic Toxicity Study in Rats. Int J Toxicol 2018; 37:327-334. [PMID: 29862856 DOI: 10.1177/1091581818777470] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Antidepressants are the most commonly prescribed drugs for psychiatric treatment, and venlafaxine (VEN) is one of the most popular options. Venlafaxine is a nontricyclic dual-acting serotonin-norepinephrine reuptake inhibitor. Although an increased incidence of acute toxicity and addiction has been reported, controlled studies examining its toxic effects on different organs are still lacking. This study investigated the possible toxic effects of VEN on the liver, kidney, and gastric tissues. Three groups of rats were administered saline, a single LD50 dose (350 mg/kg), or 100 mg/kg VEN daily, followed by increases in the dose of 50 mg/kg every 10 days for 30 days (about 10 times the therapeutic doses). The following parameters of liver and kidney injury were then assayed: alanine aminotransferase, aspartate aminotransferase, γ-glutamyl transferase, prothrombin time, partial thromboplastin time, blood urea nitrogen, and serum creatinine. A histopathological examination was then conducted. Both acute and subchronic administration of VEN produced multiple clinical manifestations in the experimental animals, including seizures, coma, and even death. Moreover, the liver and renal function tests indicated injury in these tissues. Furthermore, the histopathological examination showed signs of organ toxicity after both acute and chronic VEN exposure. This study has shown that VEN has harmful effects on the liver, kidney, and stomach in either a single high dose (LD50) or repeated exposure to 10 times the therapeutic doses. As a result, strategies to increase awareness of these effects among physicians and the public are needed because this drug may be addictive.
Collapse
Affiliation(s)
- Melad G Paulis
- 1 Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Minia University, Menia, Egypt
| | - Essam M Hafez
- 1 Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Minia University, Menia, Egypt
| | - Nashwa F El-Tahawy
- 2 Department of Histology and Cell Biology, Faculty of Medicine, Minia University, Menia, Egypt
| | - Mohmed K M Aly
- 3 Psychiatry Department, Faculty of Medicine, Minia University, Menia, Egypt
| |
Collapse
|
39
|
Applications of Mesenchymal Stem Cells in Sinus Lift Augmentation as a Dental Implant Technology. Stem Cells Int 2018; 2018:3080139. [PMID: 29760723 PMCID: PMC5926478 DOI: 10.1155/2018/3080139] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 02/13/2018] [Accepted: 02/27/2018] [Indexed: 12/26/2022] Open
Abstract
The potential application of stem cell biology in human dentistry is a new and emerging field of research. The objective of the current review was to study the efficiency of mesenchymal stem cells (MSCs) in sinus lift augmentation (SLA). A literature review was performed in PubMed Central using MeSH keywords such as sinus lift, MSCs, dental implants, and augmentation. The searches involved full-text papers written in English, published in the past 10 years (2007–2017). The review included in vitro and in vivo studies on the use of MSCs in SLA. Electronic searching provided 45 titles, and among them, 8 papers were chosen as suitable based on the inclusion requirements of this review. The reviewed studies have revealed the potential of MSCs in SLA. According to these papers, stem cell therapy combined with different biomaterials may considerably improve bone regeneration in previous steps of dental implantation and may veritably lead to efficient clinical usages in the recent future. However, the identification of an ideal source of stem cells as well as long-term studies is vital to assess the success rate of this technology. Further clinical trials are also needed to approve the potential of MSCs in SLA.
Collapse
|
40
|
Ahmadian E, Khosroushahi AY, Eftekhari A, Farajnia S, Babaei H, Eghbal MA. Novel angiotensin receptor blocker, azilsartan induces oxidative stress and NFkB-mediated apoptosis in hepatocellular carcinoma cell line HepG2. Biomed Pharmacother 2018; 99:939-946. [PMID: 29710494 DOI: 10.1016/j.biopha.2018.01.117] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 01/13/2018] [Accepted: 01/24/2018] [Indexed: 12/21/2022] Open
Abstract
Overexpression of renin angiotensin system (RAS) components and nuclear factor-kappa B (NF-kB) has a key role in various cancers. Blockade of RAS and NF-kB pathway has been suggested to reduce cancer cell proliferation. This study aimed to investigate the role of angiotensin II and NF-kB pathway in liver hepatocellular carcinoma cell line (HepG2) proliferation by using azilsartan (as a novel Ag II antagonist) and Bay 11-7082 (as NF-kB inhibitor). HepG2 cells were treated with different concentrations of azilsartan and Bay 11-7082. Cytotoxicity was determined after 24, 48, and 72?h by MTT assay. Reactive oxygen spices (ROS) generation and cytochrome c release were measured following azilsartan and Bay11- 7082 treatment. Apoptosis was analyzed qualitatively by DAPI staining and quantitatively through flow cytometry methodologies and Bax and Bcl-2 mRNA and protein levels were assessed by real time PCR and ELISA methods, respectively. The cytotoxic effects of different concentration of azilsartan and Bay11- 7082 on HepG2 cells were observed as a reduction in cell viability, increased ROS formation, cytochrome c release and apoptosis induction. These effects were found to correlate with a shift in Bax level and a downward trend in the expression of Bcl-2. These findings suggest that azilsartan and Bay11- 7082 in combination or alone have strong potential as an agent for prevention or treatment of liver cancer after further studies.
Collapse
Affiliation(s)
- Elham Ahmadian
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmacology and Toxicology Department, Maragheh University of Medical Sciences, Maragheh, Iran; Students' Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Basic Sciences, Maragheh University of Medical Sciences, Maragheh, Iran.
| | - Ahmad Yari Khosroushahi
- Department of Pharmacognosy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Aziz Eftekhari
- Pharmacology and Toxicology Department, Maragheh University of Medical Sciences, Maragheh, Iran; Department of Basic Sciences, Maragheh University of Medical Sciences, Maragheh, Iran; Toxicology Research Center, Maragheh University of Medical Sciences, Maragheh, Iran.
| | - Safar Farajnia
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Hossein Babaei
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaclogy and Toxicology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Ali Eghbal
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaclogy and Toxicology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
41
|
Eftekhari A, Ahmadian E, Azami A, Johari-Ahar M, Eghbal MA. Protective effects of coenzyme Q10 nanoparticles on dichlorvos-induced hepatotoxicity and mitochondrial/lysosomal injury. ENVIRONMENTAL TOXICOLOGY 2018; 33:167-177. [PMID: 29143438 DOI: 10.1002/tox.22505] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/22/2017] [Accepted: 10/22/2017] [Indexed: 06/07/2023]
Abstract
Development of biocompatible antioxidant nanoparticles for xenobiotic-induced liver disease treatment by oral or parenteral administration is of great interest in medicine. In the current study, we demonstrate the protective effects of coenzyme Q10 nanoparticles (CoQ10-NPs) on hepatotoxicity induced by dichlorvos (DDVP) as an organophosphate. Although CoQ10 is an efficient antioxidant, its poor bioavailability has limited the applications of this useful agent. First, CoQ10-NPs were prepared then characterized using dynamic light scattering (DLS) and transmission electron microscopy (TEM). In DDVP-treated and non-treated hepatocytes in the presence of CoQ10-NPs, cell viability, the level of reactive oxygen species (ROS), lipid peroxidation (LPO), mitochondrial membrane potential (MMP), lysosome membrane integrity, and cellular glutathione (GSH) content were measured. The prepared CoQ10-NPs were mono-dispersed and had narrow size distribution with average diameter of 54 nm. In the in vivo study, we evaluated the enzymes, which are involved in the antioxidant system for maintenance of normal liver function. In comparison to nonparticulate CoQ10, the CoQ10-NPs efficiently decreased the ROS formation, lipid peroxidation and cell death. Also, particulate form of CoQ10 improved MMP, GSH level and lysosome membrane integrity. In the in vivo, study, we revealed that CoQ10-NPs were better hepatoprotective than its nonparticulate form (P < .05). Altogether, we propose that the CoQ10-NPs have potential capability to be used as a therapeutic and prophylactic agent for poisoning that is induced by organophosphate agents, especially in the case of DDVP. Furthermore, these positive remarks make this nanoparticle amenable for the treatment of xenobiotic-induced liver diseases.
Collapse
Affiliation(s)
- Aziz Eftekhari
- Department of Basic Sciences, Maragheh university of Medical Sciences, Maragheh, Iran
- Department of Pharmacology and Toxicology, Maragheh University of Medical Sciences, Maragheh, Iran
- Toxicology Research Center, Maragheh University of Medical Sciences, Maragheh, Iran
- Managerial Epidemiology Research Center, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Elham Ahmadian
- Department of Pharmacology and Toxicology, Maragheh University of Medical Sciences, Maragheh, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aida Azami
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Johari-Ahar
- Department of Medicinal Chemistry, School of Pharmacy, Ardabil University of Medical Science, Ardabil, Iran
| | - Mohammad Ali Eghbal
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Tabriz University of Medical Sciences, Iran
| |
Collapse
|
42
|
Tiefensee Ribeiro C, Gasparotto J, Teixeira AA, Portela LVC, Flores VNL, Moreira JCF, Gelain DP. Immune neutralization of the receptor for advanced glycation end products reduce liver oxidative damage induced by an acute systemic injection of lipopolysaccharide. J Biochem 2018; 163:515-523. [DOI: 10.1093/jb/mvy013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 12/26/2017] [Indexed: 02/07/2023] Open
Affiliation(s)
| | | | | | - Luis Valmor Cruz Portela
- Laboratório de Neurotrauma e Biomarcadores, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600–Anexo, CEP 90035-003 Porto Alegre, Rio Grande do Sul, Brazil
| | | | | | | |
Collapse
|
43
|
Ahmadian E, Khosroushahi AY, Eghbal MA, Eftekhari A. Betanin reduces organophosphate induced cytotoxicity in primary hepatocyte via an anti-oxidative and mitochondrial dependent pathway. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2018; 144:71-78. [PMID: 29463411 DOI: 10.1016/j.pestbp.2017.11.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/03/2017] [Accepted: 11/29/2017] [Indexed: 06/08/2023]
Abstract
Organophosphates (OP) are potent pesticide commonly utilized in agricultural and domestic use. However, plentitude of data represent their side effects in different body tissues. We attempted to study whether betanin (a natural pigment) is able to mitigate some OPs-induced hepatotoxicity in primary rat hepatocytes. Cell viability, lactate dehydrogenase (LDH) leakage, reactive oxygen species (ROS) formation, lipid peroxidation (LPO), glutathione (GSH) depletion and mitochondrial depolarization were tested as toxicity markers. The outcomes revealed that betanin (25μM) significantly increased cell viability, plummeted ROS formation and LPO, restored cellular GSH reservoirs and protected mitochondria after chlorpyrifos (CPF) (300μM), diazinon (DZN) (600μM) and dichlrovos (DDVP) (400μM) treatment. Taken together, all data suggests the potential protective role of betanin in OPs-induced hepatotoxicity in which the mechanism appears to be inhibition of ROS formation and mitochondrial protection.
Collapse
Affiliation(s)
- Elham Ahmadian
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmacology and Toxicology Department, Maragheh University of Medical Sciences, Maragheh, Iran; Toxicology Research Center, , Maragheh University of Medical Sciences, Maragheh, Iran
| | - Ahmad Yari Khosroushahi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmacognosy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Ali Eghbal
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmacology and Toxicology Department, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Aziz Eftekhari
- Pharmacology and Toxicology Department, Maragheh University of Medical Sciences, Maragheh, Iran; Toxicology Research Center, , Maragheh University of Medical Sciences, Maragheh, Iran; Department of Basic Sciences, Maragheh University of Medical Sciences, Maragheh, Iran; Managerial Epidemiology Research Center, Maragheh University of Medical Sciences, Maragheh, Iran.
| |
Collapse
|
44
|
Eftekhari A, Ahmadian E, Azarmi Y, Parvizpur A, Khalili Fard J, Eghbal MA. Mechanistic Approach for Thioridazine-Induced Hepatotoxicity and Potential Benefits of Melatonin and/or Coenzyme Q10 on Freshly Isolated Rat Hepatocytes. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2018; 17:1465-1475. [PMID: 30568704 PMCID: PMC6269589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Thioridazine (TZ) is used mainly in the treatment of schizophrenia. However, hepatotoxicity as a life-threatening adverse effect is associated with its clinical use. In this context, we examined the cytotoxic mechanisms of TZ on freshly isolated rat hepatocytes to better understanding of the pathogenesis of TZ-induced hepatotoxicity. Hepatocytes were prepared by the method of collagenase enzyme perfusion via the portal vein. The level of parameters such as cell death, reactive oxygen species (ROS) formation, lipid peroxidation (LPO), mitochondrial membrane potential (MMP), lysosomal membrane integrity and cellular glutathione (GSH) content in TZ-treated and non-treated hepatocytes were determined and the mentioned markers were assessed in the presence of Coenzyme Q10 and/or melatonin. Results showed that TZ caused an increase in ROS formation as well as induction of LPO and GSH depletion. Moreover, mitochondria and lysosomes seem to be targets of TZ-induced toxicity. The administration of Coenzyme Q10 and/or melatonin efficiently decreased the rate of ROS formation, LPO and improved cell viability, MMP, GSH level and lysosome membrane integrity. This study proposes the possible protective role of Coenzyme Q10 and/or melatonin against TZ-induced cellular injury probably through their radical scavenging properties and their effects on mitochondria and lysosomes.
Collapse
Affiliation(s)
- Aziz Eftekhari
- Department of Pharmacology and Toxicology, Maragheh University of Medical Sciences, Maragheh, Iran.
- Toxicology Research Center, Maragheh University of Medical Sciences, Maragheh, Iran.
| | - Elham Ahmadian
- Department of Pharmacology and Toxicology, Maragheh University of Medical Sciences, Maragheh, Iran.
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Students’ Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Yadollah Azarmi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Pharmacology and Toxicology, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Alireza Parvizpur
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Pharmacology and Toxicology, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Javad Khalili Fard
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Pharmacology and Toxicology, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Ali Eghbal
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Pharmacology and Toxicology, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
45
|
Zhang Q, Liu Y, Wang H, Ma L, Xia H, Niu J, Sun T, Zhang L. The preventive effects of taurine on neural tube defects through the Wnt/PCP-Jnk-dependent pathway. Amino Acids 2017; 49:1633-1640. [PMID: 28718066 DOI: 10.1007/s00726-017-2462-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 07/03/2017] [Indexed: 11/30/2022]
Abstract
The aim of this study was to clarify the protective role of taurine in neuronal apoptosis and the role of the Wnt/PCP-Jnk pathway in mediating the preventive effects of taurine on neural tube defects (NTDs). HT-22 cells (a hippocampal neuron cell line) were divided into a control group, a glutamate-induced apoptosis group, and glutamate (4.0 mmol/L) plus low-dose taurine (L; 0.5 mmol/L) and high-dose taurine (H; 2.0 mmol/L) groups. The MTT assay was used to monitor cell proliferation and cell survival. Immunofluorescence and Western blot analyses were used to determine caspase 9 expression. Retinoic acid (RA) induced embryonic NTDs in Kunming mice, thus establishing an NTD model. Pregnant mice were divided into a control group, an RA (30 mg/kg body weight) group, and an RA (30 mg/kg body weight) plus taurine (free drinking of 2 g/L solution) group. Immunohistochemistry and Western blot analyses were used to detect the expression of Dvl, RhoA and phosphorylated (p)-Jnk/Jnk in the embryonic neural tubes. In HT-22 cells, the apoptosis rate was significantly higher and caspase 9 activation was also significantly increased in the glutamate-induced apoptosis group compared to the L and H taurine groups. In the NTD model, the expression levels of Dvl, RhoA, and p-Jnk were significantly higher in the RA group than in the control group, whereas they were significantly reduced in the RA + taurine group. This study suggests that taurine has positive effects on neuronal protection and NTD prevention. Moreover, the Wnt/PCP-Jnk-dependent pathway plays an important role in taurine-mediated prevention of NTDs.
Collapse
Affiliation(s)
- Qinghua Zhang
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Ningxia Key Laboratory of Cerebrocranial Diseases, No. 804 Shenglinajie, XingQing District, Yinchuan, 750004, NingXia, China.
| | - Yang Liu
- Department of Neurosurgery, Qingdao Chengyang People's Hospital, Qingdao, 266106, China
| | - Hui Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Monogenic Disease Research Center for Neurological Disorders, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Li Ma
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Ningxia Key Laboratory of Cerebrocranial Diseases, No. 804 Shenglinajie, XingQing District, Yinchuan, 750004, NingXia, China
| | - Hechun Xia
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Ningxia Key Laboratory of Cerebrocranial Diseases, No. 804 Shenglinajie, XingQing District, Yinchuan, 750004, NingXia, China
| | - Jianguo Niu
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Ningxia Key Laboratory of Cerebrocranial Diseases, No. 804 Shenglinajie, XingQing District, Yinchuan, 750004, NingXia, China
| | - Tao Sun
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Ningxia Key Laboratory of Cerebrocranial Diseases, No. 804 Shenglinajie, XingQing District, Yinchuan, 750004, NingXia, China
| | - Li Zhang
- Department of Cardiac Function Examination, General Hospital of Ningxia Medical University, Yinchuan, China.
| |
Collapse
|