1
|
Guo K, van den Beucken T. Advances in drug-induced liver injury research: in vitro models, mechanisms, omics and gene modulation techniques. Cell Biosci 2024; 14:134. [PMID: 39488681 PMCID: PMC11531151 DOI: 10.1186/s13578-024-01317-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024] Open
Abstract
Drug-induced liver injury (DILI) refers to drug-mediated damage to the structure and function of the liver, ranging from mild elevation of liver enzymes to severe hepatic insufficiency, and in some cases, progressing to liver failure. The mechanisms and clinical symptoms of DILI are diverse due to the varying combination of drugs, making clinical treatment and prevention complex. DILI has significant public health implications and is the primary reason for post-marketing drug withdrawals. The search for reliable preclinical models and validated biomarkers to predict and investigate DILI can contribute to a more comprehensive understanding of adverse effects and drug safety. In this review, we examine the progress of research on DILI, enumerate in vitro models with potential benefits, and highlight cellular molecular perturbations that may serve as biomarkers. Additionally, we discuss omics approaches frequently used to gather comprehensive datasets on molecular events in response to drug exposure. Finally, three commonly used gene modulation techniques are described, highlighting their application in identifying causal relationships in DILI. Altogether, this review provides a thorough overview of ongoing work and approaches in the field of DILI.
Collapse
Affiliation(s)
- Kaidi Guo
- Department of Toxicogenomics, GROW - Research Institute for Oncology & Reproduction, Maastricht University, Maastricht, 6200, MD, The Netherlands.
| | - Twan van den Beucken
- Department of Toxicogenomics, GROW - Research Institute for Oncology & Reproduction, Maastricht University, Maastricht, 6200, MD, The Netherlands
| |
Collapse
|
2
|
Wu X, Guan Y, Wang J, Song L, Zhang Y, Wang Y, Li Y, Qin L, He Q, Zhang T, Long B, Ji L. Co-catalpol alleviates fluoxetine-induced main toxicity: Involvement of ATF3/FSP1 signaling-mediated inhibition of ferroptosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 126:155340. [PMID: 38401490 DOI: 10.1016/j.phymed.2024.155340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 12/15/2023] [Accepted: 01/05/2024] [Indexed: 02/26/2024]
Abstract
BACKGROUND Fluoxetine is often used as a well-known first-line antidepressant. However, it is accompanied with hepatogenic injury as its main organ toxicity, thereby limiting its application despite its superior efficacy. Fluoxetine is commonly traditionally used combined with some Chinese antidepressant prescriptions containing Rehmannia glutinosa (Dihuang) for depression therapy and hepatoprotection. Our previous experiments showed that co-Dihuang can alleviate fluoxetine-induced liver injury while efficiencies, and catalpol may be the key ingredient to characterize the toxicity-reducing and synergistic effects. However, whether co-catalpol can alleviate fluoxetine-induced liver injury and its toxicity-reducing mechanism remain unclear. PURPOSE On the basis of the first recognition of the dose and duration at which pre-fluoxetine caused hepatic injury, co-catalpol's alleviation of fluoxetine-induced hepatic injury and its pathway was comprehensively elucidated. METHOD AND RESULTS The hepatoprotection of co-catalpol was evaluated by serum biochemical indexes sensitive to hepatic injury and multiple staining techniques for hepatic pathologic analysis. Subsequently, the pathway by which catalpol alleviated fluoxetine-induced hepatic injury was predicted by network pharmacology to be predominantly the inhibition of ferroptosis. These were validated and confirmed in subsequent experiments with key technologies and diagnostic reagents related to ferroptosis. Further molecular docking showed that activating transcription factor 3 (ATF3) and ferroptosis suppressor protein 1 (FSP1) were the the most prospective molecules for catalpol and fluoxetine among many ferroptosis-related molecules. The critical role of ATF3/FSP1 signaling was further observed by surface plasmon resonance, diagnostic reagents, transmission electron microscopy, Western blot, real-time PCR, immunofluorescence, and immunohistochemistry. Results showed that fluoxetine directly bound to ATF3 and FSP1; agonisting ATF3 or blocking FSP1 abolished the alleviation of catalpol on fluoxetine-induced liver injury, and both exacerbated ferroptosis. Moreover, co-catalpol significantly enhanced the antidepressant efficacy of fluoxetine against depressive behaviours in mice. CONCLUSION The hepatic impairment properties of fluoxetine were largely dependent on ATF3/FSP1 target-mediated ferroptosis. Co-catalpol alleviated fluoxetine-induced hepatic injury while enhancing its antidepressant efficacy, and that ATF3/FSP1 signaling-mediated inhibition of ferroptosis was involved in its co-administration detoxification mechanism. This study was the first to reveal the hepatotoxicity characteristics, targets, and mechanisms of fluoxetine; provide a detoxification and efficiency regimen by co-catalpol; and elucidate the detoxification mechanism.
Collapse
Affiliation(s)
- Xiaohui Wu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Yuechen Guan
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Junming Wang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, 450046, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, China.
| | - Lingling Song
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Yueyue Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Yanmei Wang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Yamin Li
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Lingyu Qin
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Qingwen He
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Tianzhu Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Bingyu Long
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Lijie Ji
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| |
Collapse
|
3
|
Shaer DFE, Halim HIAE. The Possible Ameliorating Role of Fisetin on Hepatic Changes Induced by Fluoxetine in Adult Male Albino Rats: Histological, Immunohistochemical, and Biochemical Study. J Microsc Ultrastruct 2023; 11:161-171. [PMID: 38025186 PMCID: PMC10679833 DOI: 10.4103/jmau.jmau_84_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 02/09/2023] Open
Abstract
Background Fluoxetine (FLX) is one of the selective serotonin reuptake inhibitors, it is widely used to treat neuropsychiatric disorders including depression, but high doses can cause several adverse effects. Fisetin (FIS), a bioactive flavonoid presents in vegetables and fruits, has antioxidant, anti-inflammatory, and anticancer effects. Aim To evaluate the possible ameliorating effect of FIS on the hepatic alterations induced by FLX in adult male albino rats. Materials and Methods Our study was done, for 3-weeks, on 48 rats that were divided into four groups: Group I (control), Group II received FIS orally (100 mg/kg/day), Group III received FLX orally (10 mg/kg/day), and Group IV concomitantly received FLX and FIS at the same dose and manner of groups II and III. Blood and liver samples were obtained and prepared for histological, immunohistochemical, and biochemical studies. Results FLX group revealed disturbed liver architecture, hepatocytes with vacuolated cytoplasm, inflammatory cellular infiltration, blood extravasation, and congestion of blood vessels in addition to, a significant increase in the area percentage of caspase-3, inducible nitric oxide synthase and the number of glial fibrillary acidic protein-expressing cells as well as a significant decrease in the area percentage of periodic acid-Schiff stain. Moreover, FLX significantly increased aspartate-aminotransferase and alanine-aminotransferase levels in the serum. In addition, FLX increased malondialdehyde level and decreased superoxide dismutase, glutathione (GSH) peroxidase, and reduced GSH levels in liver tissue. The concomitant administration of FIS ameliorated these alterations. Conclusions Administration of FIS ameliorated the histological, immunohistochemical, and biochemical alterations induced by FLX in the liver of adult male albino rats.
Collapse
Affiliation(s)
- Dina Fouad El Shaer
- Department of Histology and Cell Biology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | | |
Collapse
|
4
|
Ganguly R, Kumar R, Pandey AK. Baicalin provides protection against fluoxetine-induced hepatotoxicity by modulation of oxidative stress and inflammation. World J Hepatol 2022; 14:729-743. [PMID: 35646277 PMCID: PMC9099103 DOI: 10.4254/wjh.v14.i4.729] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/17/2021] [Accepted: 03/27/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Fluoxetine is one of the most widely prescribed anti-depressant drugs belonging to the category of selective serotonin reuptake inhibitors. Long-term fluoxetine treatment results in hepatotoxicity. Baicalin, a natural compound obtained from the Chinese herb Scutellaria baicalensis is known to have antioxidant, hepatoprotective and anti-inflammatory effects. However, the beneficial effects of baicalin against fluoxetine-induced hepatic damage have not previously been reported.
AIM To evaluate the protective action of baicalin in fluoxetine-induced liver toxicity and inflammation.
METHODS Male albino Wistar rats were divided into seven groups. Group 1 was the normal control. Oral fluoxetine was administered at 10 mg/kg body weight to groups 2, 3, 4 and 5. In addition, groups 3 and 4 were also co-administered oral baicalin (50 mg/kg and 100 mg/kg, respectively) while group 5 received silymarin (100 mg/kg), a standard hepatoprotective compound for comparison. Groups 6 and 7 were used as a positive control for baicalin (100 mg/kg) and silymarin (100 mg/kg), respectively. All treatments were carried out for 28 d. After sacrifice of the rats, biomarkers of oxidative stress [superoxide dismutase (SOD), catalase (CAT), reduced glutathione (GSH), glutathione-S-transferase (GST), advanced oxidation protein products (AOPP), malondialdehyde (MDA)], and liver injury [alanine transaminase (ALT), aspartate transaminase (AST), alkaline phosphatase (ALP), total protein, albumin, bilirubin] were studied in serum and tissue using standard protocols and diagnostic kits. Inflammatory markers [tumor necrosis factor (TNF-α), interleukin (IL)-6, IL-10 and interferon (IFN)-γ] in serum were evaluated using ELISA-based kits. The effect of baicalin on liver was also analyzed by histopathological examination of tissue sections.
RESULTS Fluoxetine-treated rats showed elevated levels of the serum liver function markers (total bilirubin, ALT, AST, and ALP) and inflammatory markers (TNF-α, IL-6, IL-10 and IFN-γ), with a decline in total protein and albumin levels. Biochemical markers of oxidative stress such as SOD, CAT, GST, GSH, MDA and AOPP in the liver tissue homogenate were also altered indicating a surge in reactive oxygen species leading to oxidative damage. Histological examination of liver tissue also showed degeneration of hepatocytes. Concurrent administration of baicalin (50 and 100 mg/kg) restored the biomarkers of oxidative stress, inflammation and hepatic damage in serum as well as in liver tissues to near normal levels.
CONCLUSION These findings suggested that long-term treatment with fluoxetine leads to oxidative stress via the formation of free radicals that consequently cause inflammation and liver damage. Concurrent treatment with baicalin alleviated fluoxetine-induced hepatotoxicity and liver injury by regulating oxidative stress and inflammation.
Collapse
Affiliation(s)
- Risha Ganguly
- Department of Biochemistry, University of Allahabad, Prayagraj 211002, India
| | - Ramesh Kumar
- Department of Biochemistry, University of Allahabad, Prayagraj 211002, India
| | - Abhay K Pandey
- Department of Biochemistry, University of Allahabad, Prayagraj 211002, India
| |
Collapse
|
5
|
The role of potassium channels on vasorelaxant effects of elabela in rat thoracic aorta. TURK GOGUS KALP DAMAR CERRAHISI DERGISI 2022; 30:18-25. [PMID: 35444849 PMCID: PMC8990140 DOI: 10.5606/tgkdc.dergisi.2022.22756] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/11/2021] [Indexed: 01/09/2023]
Abstract
Background This study aims to investigate the roles of potassium channel subtypes in the vasorelaxant effect mechanism of elabela, which is a recently discovered endogenous apelin receptor ligand. Methods The vascular rings (4-mm) obtained from the thoracic aortas of 20 male Wistar Albino rats were placed into the isolated tissue bath system. The resting tension was set to 1 g. The aortic rings were contracted with 10-5 molar phenylephrine after the equilibration period (90 min). Elabela was applied cumulatively (10-10-10-6 molar) to the aortic rings in the plateau phase. The experimental protocol was repeated in the presence of specific potassium channel subtype inhibitors to determine the role of potassium channels in the vasorelaxant effect mechanism of elabela. Results Elabela induced a concentration-dependent vasorelaxation (p<0.001). The maximum relaxation level was approximately 51% according to phenylephrineinduced contraction. Vasorelaxant effect level of elabela statistically significantly decreased after removal of the endothelium (p<0.05). Tetraethylammonium (1 milimolar), 4-Aminopyridine (1 milimolar), glyburide (10 micromolar), and barium chloride (30 micromolar) statistically significantly decreased the vasorelaxant effect level of elabela (p<0.001, p<0.001, p<0.01, and p<0.05 respectively). However, anandamide (10 micromolar) and apamin (100 nanomolar) did not statistically significantly change the vasorelaxant effect level of elabela. Conclusion Our results suggest that large-conductance calciumactivated, voltage-gated, adenosine triphosphate-sensitive, and inward-rectifier potassium channels are involved in the vasorelaxant effect mechanism of elabela in the rat thoracic aorta.
Collapse
|
6
|
Mohamed Kamel GA. Vinpocetine attenuates fluoxetine-induced liver damage in rats; Role of Nrf2 and PPAR-γ. Hum Exp Toxicol 2021; 40:S509-S518. [PMID: 34669537 DOI: 10.1177/09603271211051597] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Fluoxetine (FLX) has been widely used as first-line treatment in cases of depression and other neuropsychiatric disorders. Although its safety has been approved, the use of FLX was associated with liver injury and chronic liver disease. Vinpocetine (Vinpo), a nootropic drug, possesses antioxidant and anti-inflammatory effects. OBJECTIVE This study aimed to evaluate the protective effects of Vinpo on FLX-induced liver damage pointing to the role of peroxisome proliferator-activated receptor-gamma (PPAR-γ) and nuclear factor erythroid 2-related factor 2 (Nrf2). METHODS Rats were randomized to four groups: control group, Vinpo group (20 mg/kg/day; orally), FLX group (10 mg/kg/day; orally), and Vinpo + FLX group. RESULTS FLX-induced liver damage was evidenced through elevated liver function biomarkers and induced hepatic histopathological changes. Concurrent Vinpo treatment resulted in a significant decrease in hepatotoxicity biomarkers and histopathological alterations. FLX-induced oxidative stress and inflammation were attenuated by Vinpo. In addition, Vinpo attenuated the hepatic NRF2 and HO-1 levels and up-regulated PPAR-γ expression. Moreover, FLX elevated Bcl-2-associated X protein (Bax) mRNA expression and decreased B-cell lymphoma 2 (Bcl2) mRNA expression were markedly reversed by Vinpo. CONCLUSION Vinpo possesses ameliorative effects against FLX-induced liver injury in rats. This effect may be due to attenuation of oxidative stress and inflammation, in addition to upregulation of PPAR-γ expression.
Collapse
Affiliation(s)
- Gellan Alaa Mohamed Kamel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), 68820Al-Azhar University, Cairo, Egypt
| |
Collapse
|
7
|
Otan Özden F, Lütfioğlu M, Demir E, Bilgici B. Antioxidant effect of caffeic acid phenethyl ester in experimentally induced periodontitis. Clin Oral Investig 2021; 25:4959-4966. [PMID: 33770282 DOI: 10.1007/s00784-021-03805-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 01/20/2021] [Indexed: 12/18/2022]
Abstract
OBJECTIVES The aim of the present study was to evaluate the antioxidant effect of systemically administered caffeic acid phenethyl ester (CAPE) in periodontitis. MATERIALS AND METHODS Forty rats were randomly divided into four groups: control, lipopolysaccharide-induced experimental periodontitis (LPS), CAPE 5: LPS+5 μmol/kg/day CAPE, and CAPE 10: LPS+10 μmol/kg/day CAPE. Following lipopolysaccharide-induced experimental periodontitis, CAPE was administered intraperitoneally for 28 days. Gingival and serumal total antioxidant status (TAS) and total oxidant status (TOS) were analyzed by enzyme-linked immunosorbent assay (ELISA). RESULTS Gingival tissue TAS was significantly higher with CAPE application compared with the LPS group and was highest in the CAPE 10 group (p<0.05). Gingival tissue TOS was highest in the LPS group, and both of the CAPE dosages decreased the gingival tissue TOS, with the highest decrease in the CAPE 10 group (p<0.05). The differences were not significant for serumal TAS or TOS levels (p>0.05). CONCLUSIONS The effect of CAPE on increased TAS and decreased TOS levels in inflamed gingival tissue indicates the antioxidant therapeutic potential of CAPE in periodontitis. CLINICAL RELEVANCE Within the limitations of this study, CAPE may be suggested as an effective host modulator agent for reducing oxidative stress in gingival tissue and might be considered as an adjunctive therapy in periodontitis.
Collapse
Affiliation(s)
- Feyza Otan Özden
- Department of Periodontology, School of Dentistry, Ondokuz Mayıs University, Kurupelit, 55139, Samsun, Turkey.
| | - Müge Lütfioğlu
- Department of Periodontology, School of Dentistry, Ondokuz Mayıs University, Kurupelit, 55139, Samsun, Turkey
| | - Esra Demir
- Department of Periodontology, School of Dentistry, Biruni University, İstanbul, Turkey
| | - Birşen Bilgici
- Department of Biochemistry, School of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| |
Collapse
|
8
|
Grzegorzewska AE, Adamska P, Iwańczyk-Skalska E, Ostromecka K, Niepolski L, Marcinkowski W, Mostowska A, Warchoł W, Żaba C, Jagodziński PP. Paraoxonase 1 concerning dyslipidaemia, cardiovascular diseases, and mortality in haemodialysis patients. Sci Rep 2021; 11:6773. [PMID: 33762698 PMCID: PMC7990965 DOI: 10.1038/s41598-021-86231-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 02/23/2021] [Indexed: 01/31/2023] Open
Abstract
Paraoxonase 1 (PON1) is known for preventing atherosclerosis through lipid-modifying features, antioxidant activity, anti-inflammatory, anti-apoptosis, anti-thrombosis, and anti-adhesion properties. Uremic patients requiring haemodialysis (HD) are especially prone to atherosclerosis and its complications. We analysed the PON1 gene (PON1) polymorphisms and serum PON1 (paraoxonase) activity concerning dyslipidaemia and related cardiovascular diseases and mortality to show how they associate under uremic conditions modified by maintenance HD treatment. The rs662 AA + AG (OR 1.76, 95%CI 1.10-2.80, P = 0.018), rs854560 TT (OR 1.48, 95%CI 1.04-2.11, P = 0.031), and rs854560 AT + TT (OR 1.28, 95%CI 1.01-1.63, P = 0.040) contributed to the prevalence of atherogenic dyslipidaemia diagnosed by the triglyceride (TG)/HDL-cholesterol ratio ≥ 3.8. The normalized serum PON1 activity positively correlated with atherogenic dyslipidaemia (ẞ 0.67 ± 0.25, P = 0.008). The PON1 rs854560 allele T was involved in the higher prevalence of ischemic cerebral stroke (OR 1.38, 1.02-1.85, P = 0.034). The PON1 rs705379 TT genotype contributed to cardiovascular (HR 1.27, 95% CI 1.03-1.57, P = 0.025) and cardiac (HR 1.34, 95% CI 1.05-1.71, P = 0.018) mortality. All P-values were obtained in multiple regression analyses, including clinical variables. Multifaceted associations of PON1 with dyslipidaemia, ischemic cerebral stroke, and cardiovascular mortality in HD patients provide arguments for the consideration of PON1 and its protein product as therapeutic targets in the prevention of atherosclerosis and its complications in uremic patients.
Collapse
Affiliation(s)
- Alicja E. Grzegorzewska
- grid.22254.330000 0001 2205 0971Department of Nephrology, Transplantology and Internal Diseases, Poznan University of Medical Sciences, Przybyszewskiego 49, 60-355 Poznań, Poland
| | - Paulina Adamska
- grid.22254.330000 0001 2205 0971Department of Nephrology, Transplantology and Internal Diseases, Poznan University of Medical Sciences, Przybyszewskiego 49, 60-355 Poznań, Poland
| | - Ewa Iwańczyk-Skalska
- grid.22254.330000 0001 2205 0971Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 60-781 Poznań, Poland
| | - Kamila Ostromecka
- grid.22254.330000 0001 2205 0971Nephrology Research Group, Department of Nephrology, Transplantology and Internal Diseases, Poznan University of Medical Sciences, 60-355 Poznań, Poland
| | - Leszek Niepolski
- B. Braun Avitum Poland, Dialysis Center, 64-300 Nowy Tomyśl, Poland
| | | | - Adrianna Mostowska
- grid.22254.330000 0001 2205 0971Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 60-781 Poznań, Poland
| | - Wojciech Warchoł
- grid.22254.330000 0001 2205 0971Department of Ophthalmology and Optometry, Poznan University of Medical Sciences, 60-806 Poznań, Poland
| | - Czesław Żaba
- grid.22254.330000 0001 2205 0971Department of Forensic Medicine, Poznan University of Medical Sciences, 60-781 Poznań, Poland
| | - Paweł P. Jagodziński
- grid.22254.330000 0001 2205 0971Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 60-781 Poznań, Poland
| |
Collapse
|
9
|
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: 57] [Impact Index Per Article: 14.3] [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
|
10
|
Zaitone SA, Ahmed E, Elsherbiny NM, Mehanna ET, El-Kherbetawy MK, ElSayed MH, Alshareef DM, Moustafa YM. Caffeic acid improves locomotor activity and lessens inflammatory burden in a mouse model of rotenone-induced nigral neurodegeneration: Relevance to Parkinson's disease therapy. Pharmacol Rep 2018; 71:32-41. [PMID: 30368226 DOI: 10.1016/j.pharep.2018.08.004] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 08/02/2018] [Accepted: 08/10/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Caffeic acid phenethyl ester is found in honey bee propolis. It has immunomodulatory, anti-inflammatory and anti-cancer properties. Rotenone is a pesticide commonly used for inducing experimental Parkinson's disease (PD) due to complex I inhibition and microglia activating properties. The current study examined neuroprotective effect of caffeic acid against rotenone-induced neurodegeneration in groups of seven mice. METHODS Mice received protective doses of caffeic acid (2.5, 5 or 10 mg/kg) daily and nine injections of rotenone (1 mg kg, subcutaneously) - every 48 h. Behavioral evaluation of motor function was done by a battery of tests including open-field test, cylinder test, pole test and rotarod test; all these tests showed motor impairment. RESULTS Assay of striatal dopamine highlighted a significant decrease and increases in inflammatory markers. In addition, histopathological assessment of substantia nigra neurons demonstrated low immunostaining for tyrosine hydroxylase (TH) in rotenone treated mice. PCR analysis highlighted upregulation for genes encoding CD11b (a microglia surface antigen), cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS) and nuclear factor-κB (NFκB). Treatment with caffeic acid (5 or 10 mg/kg) amended most of rotenone-induced motor deficits, lessened microglia expression and inflammatory mediators and improved the nigral TH immunostaining. CONCLUSION These results confirmed the anti-inflammatory activity of caffeic acid and highlighted its neuroprotective activity against rotenone-induced neurodegeneration in mice.
Collapse
Affiliation(s)
- Sawsan A Zaitone
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt.
| | - Eman Ahmed
- Clinical Pharmacology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Nehal M Elsherbiny
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia; Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Eman T Mehanna
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | | | - Mohamed H ElSayed
- Department of Physiology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Duha M Alshareef
- Department of Pharmaceutics, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Yasser M Moustafa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| |
Collapse
|