1
|
Yoladi FB, Palabiyik-Yucelik SS, Bahador Zirh E, Halici Z, Baydar T. Effects of idebenone and coenzyme Q10 on NLRP3/caspase-1/IL-1β pathway regulation on ethanol-induced hepatotoxicity in rats. Drug Chem Toxicol 2024; 47:1205-1217. [PMID: 38804209 DOI: 10.1080/01480545.2024.2351191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 04/29/2024] [Indexed: 05/29/2024]
Abstract
Chronic and excessive alcohol consumption leads to liver toxicity. There is a need to investigate effective therapeutic strategies to alleviate alcohol-induced liver injury, which remains the leading cause of liver-related morbidity and mortality worldwide. Therefore here, we looked into and evaluated how ethanol-induced hepatotoxicity was affected by coenzyme Q10 (CoQ10) and its analog, idebenone (IDE), on the NLRP3/caspase-1/IL-1 pathway. Hepatotoxicity induced in rats through the oral administration of gradually increasing dosages of ethanol (from 2 to 6 g/kg/day) over 30 days and the effect of CoQ10 (10 or 20 mg/kg) and IDE (50 or 100 mg/kg) were evaluated. Serum hepatotoxicity markers (ALT, AST, GGT, ALP, and TBIL), tissue oxidative stress markers and the mRNA expressions of IL-1β, IL-18, TGF-β, NF-κB, NLRP3, and caspase-1 were evaluated. Masson's trichrome staining was also used to visualize fibrosis in the liver tissue. The results indicated that ethanol exposure led to hepatotoxicity as well as considerable NLRP3/caspase-1/IL-1β pathway activation. Moreover, CoQ10 or IDE treatment reduced measured parameters in a dosage-dependent manner. Thus, by inhibiting the NLRP3/caspase-1/IL-1 pathway, CoQ10 and IDE can prevent the hepatotoxicity caused by ethanol, although CoQ10 is more effective than IDE. This study will provide insight into new therapeutic avenues that take advantage of the anti-inflammatory and antioxidant properties of CoQ10 and IDE in ethanol-induced liver diseases.
Collapse
Affiliation(s)
- Fatma Betül Yoladi
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Atatürk University, Erzurum, Turkey
- Department of Toxicology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Saziye Sezin Palabiyik-Yucelik
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Atatürk University, Erzurum, Turkey
- Clinical Research, Development and Design Application and Research Center, Atatürk University, Erzurum, Turkey
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Ondokuz Mayıs University, Samsun, Turkey
| | - Elham Bahador Zirh
- Department of Histology and Embryology, Faculty of Medicine, TOBB University of Economics and Technology, Ankara, Turkey
| | - Zekai Halici
- Clinical Research, Development and Design Application and Research Center, Atatürk University, Erzurum, Turkey
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Terken Baydar
- Department of Toxicology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|
2
|
Akbel E, Kucukkurt I, Ince S, Demirel HH, Acaroz DA, Zemheri-Navruz F, Kan F. Investigation of protective effect of resveratrol and coenzyme Q 10 against cyclophosphamide-induced lipid peroxidation, oxidative stress and DNA damage in rats. Toxicol Res (Camb) 2024; 13:tfad123. [PMID: 38173543 PMCID: PMC10758596 DOI: 10.1093/toxres/tfad123] [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: 08/27/2023] [Revised: 12/05/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024] Open
Abstract
It is seen that cyclophosphamide, which is used in treating many diseases, especially cancer, causes toxicity in studies, and its metabolites induce oxidative stress. This study aimed to investigate the protective effects of resveratrol and Coenzyme Q10, known for their antioxidant properties, separately and together, against oxidative stress induced by cyclophosphamide. In this study, 35 Wistar albino male rats were divided into five groups. Groups; Control group, cyclophosphamide (CP) group (CP as 75 mg kg i.p. on day 14), coenzyme Q10 (CoQ10) + CP group (20 mg/kg i.p. CoQ10 + 75 mg kg i.p. CP), resveratrol (Res) + CP group (20 mg/kg i.p. Res + 75 mg/kg i.p. CP), CoQ10 + Res + CP group (20 mg/kg i.p Res + 20 mg/kg i.p CoQ10 and 75 mg/kg i.p.CP). At the end of the experiment, the cholesterol, creatinine and urea levels of the group given CP increased, while a decrease was observed in the groups given Res and CoQ10. Malondialdehyde level was high, glutathione level, superoxide dismutase and catalase activities were decreased in the blood and all tissues (liver, kidney, brain, heart and testis) of the CP given group. DNA damage and histopathological changes were also observed. In contrast, Res and CoQ10, both separately and together, reversed the CP-induced altered level and enzyme activities and ameliorated DNA damage and histopathological changes. In this study, the effects of Res and CoQ10 against CP toxicity were examined both separately and together.
Collapse
Affiliation(s)
- Erten Akbel
- Usak Health Training School, Usak University, 64200, Uşak, Turkey
| | - Ismail Kucukkurt
- Department of Biochemistry, Faculty of Veterinary Medicine, Afyon Kocatepe University, 03200, Afyonkarahisar, Turkey
| | - Sinan Ince
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Afyon Kocatepe University, 03200, Afyonkarahisar, Turkey
| | | | - Damla Arslan Acaroz
- Department of Biochemistry, Faculty of Veterinary Medicine, Afyon Kocatepe University, 03200, Afyonkarahisar, Turkey
| | - Fahriye Zemheri-Navruz
- Faculty of Science, Department of Molecular Biology and Genetics, Bartın University, 74110, Bartın, Turkey
| | - Fahriye Kan
- Department of Biochemistry, Faculty of Veterinary Medicine, Afyon Kocatepe University, 03200, Afyonkarahisar, Turkey
| |
Collapse
|
3
|
Hooshangi Shayesteh MR, Hami Z, Chamanara M, Parvizi MR, Golaghaei A, Nassireslami E. Evaluation of the protective effect of coenzyme Q 10 on hepatotoxicity caused by acute phosphine poisoning. Int J Immunopathol Pharmacol 2024; 38:3946320241250286. [PMID: 38764158 PMCID: PMC11104032 DOI: 10.1177/03946320241250286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 04/12/2024] [Indexed: 05/21/2024] Open
Abstract
Background: Aluminum phosphide (AlP) poisoning is prevalent in numerous countries, resulting in high mortality rates. Phosphine gas, the primary agent responsible for AlP poisoning, exerts detrimental effects on various organs, notably the heart, liver and kidneys. Numerous studies have documented the advantageous impact of Coenzyme Q10 (CoQ10) in mitigating hepatic injuries. The objective of this investigation is to explore the potential protective efficacy of CoQ10 against hepatic toxicity arising from AlP poisoning. Method: The study encompassed distinct groups receiving almond oil, normal saline, exclusive CoQ10 (at a dosage of 100 mg/kg), AlP at 12 mg/kg; LD50 (lethal dose for 50%), and four groups subjected to AlP along with CoQ10 administration (post-AlP gavage). CoQ10 was administered at 10, 50, and 100 mg/kg doses via Intraparietal (ip) injections. After 24 h, liver tissue specimens were scrutinized for mitochondrial complex activities, oxidative stress parameters, and apoptosis as well as biomarkers such as aspartate transaminase (AST) and alanine transaminase (ALT). Results: AlP induced a significant decrease in the activity of mitochondrial complexes I and IV, as well as a reduction in catalase activity, Ferric Reducing Antioxidant Power (FRAP), and Thiol levels. Additionally, AlP significantly elevated oxidative stress levels, indicated by elevated reactive oxygen species (ROS) production, and resulted in the increment of hepatic biomarkers such as AST and ALT. Administration of CoQ10 led to a substantial improvement in the aforementioned biochemical markers. Furthermore, phosphine exposure resulted in a significant reduction in viable hepatocytes and an increase in apoptosis. Co-treatment with CoQ10 exhibited a dose-dependent reversal of these observed alterations. Conclusion: CoQ10 preserved mitochondrial function, consequently mitigating oxidative damage. This preventive action impeded the progression of heart cells toward apoptosis.
Collapse
Affiliation(s)
- Mohammad Reza Hooshangi Shayesteh
- Department of Pharmacology and Toxicology, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Zahra Hami
- Department of Pharmacology and Toxicology, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Mohsen Chamanara
- Department of Pharmacology and Toxicology, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Parvizi
- Department of Physiology, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Alireza Golaghaei
- Department of Pharmacology and Toxicology, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Ehsan Nassireslami
- Department of Pharmacology and Toxicology, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Abd-Elhakim YM, Hashem MMM, Abo-El-Sooud K, Mousa MR, Soliman AM, Mouneir SM, Ismail SH, Hassan BA, El-Nour HHM. Interactive effects of cadmium and titanium dioxide nanoparticles on hepatic tissue in rats: Ameliorative role of coenzyme 10 via modulation of the NF-κB and TNFα pathway. Food Chem Toxicol 2023; 182:114191. [PMID: 37980978 DOI: 10.1016/j.fct.2023.114191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 11/06/2023] [Accepted: 11/11/2023] [Indexed: 11/21/2023]
Abstract
This study investigated the effect of oral dosing of titanium dioxide nanoparticles (TNPs) and cadmium (Cd2+) on rat liver and the potential protective role of coenzyme Q10 (CQ10) against TNPs and Cd2+-induced hepatic injury. Seventy male Sprague Dawley rats were divided into seven groups and orally given distilled water, corn oil, CQ10 (10 mg/kg b.wt), TNPs (50 mg/kg b.wt), Cd2+ (5 mg/kg b.wt), TNPs + Cd2+, or TNPs + Cd2++CQ10 by gastric gavage for 60 successive days. The results showed that individual or mutual exposure to TNPs and Cd2+ significantly increased the serum levels of various hepatic enzymes and lipids, depleted the hepatic content of antioxidant enzymes, and increased malondialdehyde. Moreover, the hepatic titanium and Cd2+ content were increased considerably in TNPs and/or Cd2+-exposed rats. Furthermore, marked histopathological perturbations with increased immunoexpression of tumor necrosis factor-alpha and nuclear factor kappa B were evident in TNPs and/or Cd2+-exposed rats. However, CQ10 significantly counteracted the damaging effect of combined exposure of TNPs and Cd2+ on the liver. The study concluded that TNPs and Cd2+ exposure harm hepatic function and its architecture, particularly at their mutual exposure, but CQ10 could be a candidate protective agent against TNPs and Cd2+ hepatotoxic impacts.
Collapse
Affiliation(s)
- Yasmina M Abd-Elhakim
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt.
| | - Mohamed M M Hashem
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Khaled Abo-El-Sooud
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Mohamed R Mousa
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Ahmed M Soliman
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Samar M Mouneir
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Sameh H Ismail
- Faculty of Nanotechnology for Postgraduate Studies, Cairo University, Sheikh Zayed Campus, 6th October City, Giza, 12588, Egypt
| | - Bayan A Hassan
- Pharmacology Department, Faculty of Pharmacy, Future University, Cairo 11835, Egypt
| | - Hayat H M El-Nour
- Biology of Reproduction Department, Animal Reproduction Research Institute, Giza 3514805, Egypt
| |
Collapse
|
5
|
Fakharaldeen ZA, Al-Mudhafar A, Gany SN, Radhi AN, Hadi NR. Neuroprotective effects of Coenzyme Q10 in ischemia-reperfusion injury via inflammation and oxidative stress reduction in adult male rats. J Med Life 2023; 16:1534-1539. [PMID: 38313176 PMCID: PMC10835568 DOI: 10.25122/jml-2023-0099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 04/28/2023] [Indexed: 02/06/2024] Open
Abstract
This study aimed to investigate the potential neuroprotective effects of coenzyme Q10 in cerebral ischemia-reperfusion injury-induced neuronal damage and explore the underlying mechanisms. Twenty-eight adult male rats, weighing approximately 200-300 grams, were randomly divided into four groups: the sham group (neck dissection without ischemia), the control group (30 minutes of bilateral common carotid artery ligation followed by one hour of reperfusion), the vehicle group (oral carboxymethylcellulose solution for seven days prior to bilateral common carotid artery ligation and reperfusion), and the treatment group (seven days of coenzyme Q10 pretreatment followed by bilateral common carotid artery occlusion and reperfusion). Histopathological analysis and measurement of brain infarct size were performed, and cerebral levels of IL-6, IL-10, TNF-α, ICAM-1, NF-κB p65, and total antioxidant capacity were assessed. These cerebral tissue levels and cerebral infarct size were significantly elevated in the control and vehicle groups compared to the sham group. Conversely, the total antioxidant capacity was significantly reduced in these groups. Coenzyme Q10 treatment resulted in a significant increase in IL-10 and total antioxidant capacity levels, along with a significant decrease in IL-6, ICAM-1, TNF-α, and NF-κB p65 levels. Histopathological analysis revealed a significant reduction in ischemic damage in the coenzyme Q10-treated group. Coenzyme Q10 has neuroprotective properties in rats subjected to cerebral ischemia/reperfusion injury, possibly through its anti-inflammatory and anti-oxidative effects.
Collapse
Affiliation(s)
- Zainab Ali Fakharaldeen
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Najaf, Iraq
| | - Ahmed Al-Mudhafar
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Najaf, Iraq
| | - Sarmad Nory Gany
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Najaf, Iraq
| | | | - Najah Rayish Hadi
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Najaf, Iraq
| |
Collapse
|
6
|
da Silva RHS, de Moura M, de Paula L, Arantes KC, da Silva M, de Amorim J, Miguel MP, Martins DB, de Melo e Silva D, Melo MM, Botelho AFM. Effects of coenzyme Q10 and N-acetylcysteine on experimental poisoning by paracetamol in Wistar rats. PLoS One 2023; 18:e0290268. [PMID: 37607187 PMCID: PMC10443853 DOI: 10.1371/journal.pone.0290268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 08/05/2023] [Indexed: 08/24/2023] Open
Abstract
Paracetamol (PAR) is a drug widely used in human and veterinary medicine as an analgesic and antipyretic, often involved in cases of intoxication. The most common clinical signs result from damage to red blood cells and hepatocytes, and this intoxication is considered a model for the induction of acute liver failure. In the present study, the hepatoprotective effects of coenzyme Q10 (CoQ10) and N-acetylcysteine (NAC) against experimental paracetamol (PAR) poisoning were analysed. Thirty-five adult Wistar rats (Rattus novergicus albinus) were randomly assigned to five groups, and thirty-one of these survived the treatments. Negative control group (CON-) received 1mL of 0.9% NaCl orally (PO). Other groups received 1.2g/kg of PAR (PO). Positive control group (CON+) received only PAR. NAC group received 800 mg/kg intraperitoneally (IP) of NAC 1h after the administration of PAR and at 12 h received 1mL of 0.9% NaCl, IP. The fourth group (CoQ10) received 1h and 12 h after intoxication, CoQ10 (10mg/kg IP). And the fifth group (NAC+CoQ10) received NAC (800mg/kg, IP) and CoQ10 (10mg/kg, IP). After 12 hours, the rats were euthanized and necropsied to collect liver and kidney tissues for histopathological evaluation and electronic microscopy. A single dose of PAR caused severe acute hepatitis. NAC couldn't reverse the liver and kidney damages. The group that received CoQ10 and NAC had moderate liver damage, while the group that received only CoQ10 had lower values of liver enzymes and mild liver and kidney damage. Animals that received treatment with CoQ10 or NAC+CoQ10 presented normal hepatocyte mitochondria and nuclei. Although CoQ10 couldn't reverse PAR organ damage, results indicate promising hepatoprotection in Wistar rats.
Collapse
Affiliation(s)
- Rayanne Henrique Santana da Silva
- Veterinary Toxicology Laboratory, Veterinary Hospital, Veterinary and Animal Science School, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Mariana de Moura
- Department of Genetics, Laboratory of Mutagenesis, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Larissa de Paula
- Pathology Sector, Instituto de Patologia Tropical e Saúde Pública, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Kelly Carolina Arantes
- Veterinary Toxicology Laboratory, Veterinary Hospital, Veterinary and Animal Science School, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Marina da Silva
- Veterinary Clinical Pathology Laboratory, Veterinary Hospital, Veterinary and Animal Science School, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Jaqueline de Amorim
- Veterinary Clinical Pathology Laboratory, Veterinary Hospital, Veterinary and Animal Science School, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Marina Pacheco Miguel
- Pathology Sector, Instituto de Patologia Tropical e Saúde Pública, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Danieli Brolo Martins
- Veterinary Clinical Pathology Laboratory, Veterinary Hospital, Veterinary and Animal Science School, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Daniela de Melo e Silva
- Department of Genetics, Laboratory of Mutagenesis, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Marília Martins Melo
- Veterinary Toxicology Laboratory, Veterinary Hospital, Veterinary and Animal Science School, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ana Flávia Machado Botelho
- Veterinary Toxicology Laboratory, Veterinary Hospital, Veterinary and Animal Science School, Federal University of Goiás, Goiânia, Goiás, Brazil
| |
Collapse
|
7
|
Bagheri S, Haddadi R, Saki S, Kourosh-Arami M, Rashno M, Mojaver A, Komaki A. Neuroprotective effects of coenzyme Q10 on neurological diseases: a review article. Front Neurosci 2023; 17:1188839. [PMID: 37424991 PMCID: PMC10326389 DOI: 10.3389/fnins.2023.1188839] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/22/2023] [Indexed: 07/11/2023] Open
Abstract
Neurological disorders affect the nervous system. Biochemical, structural, or electrical abnormalities in the spinal cord, brain, or other nerves lead to different symptoms, including muscle weakness, paralysis, poor coordination, seizures, loss of sensation, and pain. There are many recognized neurological diseases, like epilepsy, Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), stroke, autosomal recessive cerebellar ataxia 2 (ARCA2), Leber's hereditary optic neuropathy (LHON), and spinocerebellar ataxia autosomal recessive 9 (SCAR9). Different agents, such as coenzyme Q10 (CoQ10), exert neuroprotective effects against neuronal damage. Online databases, such as Scopus, Google Scholar, Web of Science, and PubMed/MEDLINE were systematically searched until December 2020 using keywords, including review, neurological disorders, and CoQ10. CoQ10 is endogenously produced in the body and also can be found in supplements or foods. CoQ10 has antioxidant and anti-inflammatory effects and plays a role in energy production and mitochondria stabilization, which are mechanisms, by which CoQ10 exerts its neuroprotective effects. Thus, in this review, we discussed the association between CoQ10 and neurological diseases, including AD, depression, MS, epilepsy, PD, LHON, ARCA2, SCAR9, and stroke. In addition, new therapeutic targets were introduced for the next drug discoveries.
Collapse
Affiliation(s)
- Shokufeh Bagheri
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rasool Haddadi
- Department of Pharmacology, School of Pharmacy, Hamadan University of Medical Science, Hamadan, Iran
| | - Sahar Saki
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Masoumeh Kourosh-Arami
- Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Masome Rashno
- Asadabad School of Medical Sciences, Asadabad, Iran
- Student Research Committee, Asadabad School of Medical Sciences, Asadabad, Iran
| | - Ali Mojaver
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Komaki
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
8
|
Hossain M, Suchi TT, Samiha F, Islam MM, Tully FA, Hasan J, Rahman MA, Shill MC, Bepari AK, Rahman GS, Reza HM. Coenzyme Q10 ameliorates carbofuran induced hepatotoxicity and nephrotoxicity in wister rats. Heliyon 2023; 9:e13727. [PMID: 36865458 PMCID: PMC9971173 DOI: 10.1016/j.heliyon.2023.e13727] [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: 06/20/2022] [Revised: 02/03/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
Carbofuran is a widely used poisonous pesticide around the world that helps to control insects during farming. Upon oral ingestion to humans, it exaggerates oxidative stress in various organs like the liver, brain, kidney, and heart. Several studies reported that oxidative stress in the liver initiates and propagates hepatic cell necrosis, ultimately resulting in hepatotoxicity. It also reported that coenzyme Q10 (CoQ10) can neutralize oxidative stress due to its antioxidant properties. However, the hepatoprotective and nephroprotective role of CoQ10 against carbofuran toxicity has not been investigated. Therefore, the present study aimed to evaluate the hepatoprotective and nephroprotective role of CoQ10 in carbofuran-induced hepatotoxicity and nephrotoxicity in a mouse model for the first time. We determined the blood serum diagnostic markers, oxidative stress parameters, antioxidant system, and histopathological characteristics of liver and kidney tissues. The administration of 100 mg/kg of CoQ10 in carbofuran-treated rats significantly attenuated AST, ALT, ALP, serum creatinine, and BUN levels. Moreover, CoQ10 (100 mg/kg) remarkably altered the level of NO, MDA, AOPP, GSH, SOD, and CAT in both the liver and kidney. The histopathological data also unveiled that CoQ10 treatment prevented inflammatory cell infiltration in carbofuran-exposed rats. Therefore, our findings infer that CoQ10 may effectively protect liver and kidney tissues against carbofuran-induced oxidative hepatotoxicity and nephrotoxicity.
Collapse
Affiliation(s)
- Murad Hossain
- Department of Pharmaceutical Sciences, School of Health & Life Sciences, North South University, Dhaka 1229, Bangladesh,Corresponding author.
| | - Tamanna Tanjim Suchi
- Department of Pharmaceutical Sciences, School of Health & Life Sciences, North South University, Dhaka 1229, Bangladesh
| | - Farzana Samiha
- Department of Pharmaceutical Sciences, School of Health & Life Sciences, North South University, Dhaka 1229, Bangladesh
| | - M.M. Monirul Islam
- Department of Pharmaceutical Sciences, School of Health & Life Sciences, North South University, Dhaka 1229, Bangladesh
| | - Fahima Abdullah Tully
- Department of Pharmaceutical Sciences, School of Health & Life Sciences, North South University, Dhaka 1229, Bangladesh
| | - Javed Hasan
- Department of Pharmaceutical Sciences, School of Health & Life Sciences, North South University, Dhaka 1229, Bangladesh
| | - Md Ashrafur Rahman
- Department of Pharmaceutical Sciences, School of Health & Life Sciences, North South University, Dhaka 1229, Bangladesh,Department of Pharmaceutical Sciences, Wilkes university, Pennsylvania, USA, 18766
| | - Manik Chandra Shill
- Department of Pharmaceutical Sciences, School of Health & Life Sciences, North South University, Dhaka 1229, Bangladesh
| | - Asim Kumar Bepari
- Department of Pharmaceutical Sciences, School of Health & Life Sciences, North South University, Dhaka 1229, Bangladesh
| | - G.M. Sayedur Rahman
- Department of Pharmaceutical Sciences, School of Health & Life Sciences, North South University, Dhaka 1229, Bangladesh
| | - Hasan Mahmud Reza
- Department of Pharmaceutical Sciences, School of Health & Life Sciences, North South University, Dhaka 1229, Bangladesh,Corresponding author.
| |
Collapse
|
9
|
Motafeghi F, Mortazavi P, Ghassemi-Barghi N, Zahedi M, Shokrzadeh M. Dexamethasone as an anti-cancer or hepatotoxic. Toxicol Mech Methods 2023; 33:161-171. [PMID: 35866224 DOI: 10.1080/15376516.2022.2105183] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The linkage between inflammation and oxidative stress in liver damage has been proven and is undeniable; dexamethasone with some antioxidants can reduce the toxicity of liver tissue. Due to the importance of cancer treatment, glucocorticoids' synergistic effect in inhibiting cancer cell growth is also investigated. Dexamethasone alone and combined with etoposide were tested at concentrations of 1, 5, and 10 μM to evaluate the potency of dexamethasone in inhibiting the growth of A549 cells using oxidative stress factors and DNA damage. Also, intraperitoneal injection of dexamethasone in rats was used to induce liver toxicity. Coenzyme Q10 at different concentrations (1, 10, and 50 mg/kg) was used as an antioxidant to assess the oxidative stress factors and measure Caspase-3 activity. The results showed that dexamethasone combined with etoposide could significantly inhibit the growth of cancer cells and induce apoptosis. Treatment of A549 cells using dexamethasone also inhibits cancer cells' growth by inducing oxidative stress and DNA damage. Dexamethasone also, by inducing oxidative stress and activation of caspase 3, ultimately causes hepatotoxicity. Treatment with different concentrations of CoQ10 showed improved mitochondrial function, antioxidant defense, and liver enzyme. The best effect of coenzyme Q10 on dexamethasone-induced hepatotoxicity is 50 mg/kg. As a result, dexamethasone (alone and combined with etoposide) has an anti-cancer effect by damaging DNA and inducing oxidative stress. Also, CoQ10 has antioxidant effects against dexamethasone-induced hepatotoxicity by improving mitochondrial function and reducing caspase-3 activity.
Collapse
Affiliation(s)
- Farzaneh Motafeghi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Parham Mortazavi
- Department of Pharmacology and Toxicology, Student Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nasrin Ghassemi-Barghi
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Zahedi
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.,Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Shokrzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
10
|
Ugan RA, Cadirci E, Un H, Cinar I, Gurbuz MA. Fisetin Attenuates Paracetamol-Induced Hepatotoxicity by Regulating CYP2E1 Enzyme. AN ACAD BRAS CIENC 2023; 95:e20201408. [PMID: 37018834 DOI: 10.1590/0001-3765202320201408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/14/2020] [Indexed: 04/07/2023] Open
Abstract
Paracetamol is one of the drugs that cause hepatic damage. Fisetin has wide pharmacological effects such as anticancer, antiinflammatory and antioxidant. We aimed to evaluate the possible protective effect of fisetin on paracetamol-induced hepatotoxicity. Fisetin was administered at 25 and 50 mg/kg doses. Paracetamol was administered orally at a dose of 2 g/kg for induce hepatotoxicity 1 h after the fisetin and NAC treatments. The rats were sacrificed 24h after the Paracetamol administration. Tumor necrosis factor-alpha (TNF-α), NFκB and CYP2E1 mRNA levels and Superoxide dismutase (SOD) activity, glutathione (GSH) and malondialdehyde (MDA) levels of livers were determined. Serum ALT, AST and ALP levels were measured. Histopathological examinations were also performed. Fisetin administration significantly decreased the ALT, AST and ALP levels in a dose dependent manner. In addition, SOD activity and GSH levels increased, and the MDA level decreased with the treatment of fisetin. The TNF-α, NFκB and CYP2E1 gene expressions were significantly lower in both doses of the fisetin groups compared with the PARA group. Histopathological examinations showed that fisetin has hepatoprotective effects. This study showed that fisetin has the liver protective effects by increasing GSH, decreasing inflammatory mediators and CYP2E1.
Collapse
Affiliation(s)
- Rustem A Ugan
- Ataturk University, Faculty of Pharmacy, Department of Pharmacology, 25240, Erzurum, Turkey
- Ataturk University, Clinical Research, Development and Design Application and Research Center, 25240, Erzurum, Turkey
| | - Elif Cadirci
- Ataturk University, Faculty of Medicine, Department of Pharmacology, 25240, Erzurum, Turkey
- Ataturk University, Clinical Research, Development and Design Application and Research Center, 25240, Erzurum, Turkey
| | - Harun Un
- Agri Ibrahim Cecen University, Faculty of Pharmacy, Department of Biochemistry, 04100, Agri, Turkey
| | - Irfan Cinar
- Kafkas University, Faculty of Medicine, Department of Pharmacology, Kars, 36200, Turkey
| | - Muhammet A Gurbuz
- Ataturk University, Faculty of Medicine, Department of Histology and Embryology, 25240, Erzurum, Turkey
| |
Collapse
|
11
|
Rosuvastatin and co-enzyme Q10 improve high-fat and high-fructose diet-induced metabolic syndrome in rats via ameliorating inflammatory and oxidative burden. Biomed Pharmacother 2022; 153:113526. [PMID: 36076607 DOI: 10.1016/j.biopha.2022.113526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 08/04/2022] [Accepted: 08/08/2022] [Indexed: 11/17/2022] Open
|
12
|
Abdelnaby A, Abdel-Aleem N, Mansour A, Abdelkader A, Ibrahim AN, Sorour SM, Elgendy E, Bayoumi H, Abdelrahman SM, Ibrahim SF, Alsaati I, Abdeen A. The Combination of Tamarindus indica and Coenzyme Q10 can be a Potential Therapy Preference to Attenuate Cadmium-Induced Hepatorenal Injury. Front Pharmacol 2022; 13:954030. [PMID: 36003506 PMCID: PMC9393486 DOI: 10.3389/fphar.2022.954030] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
Cadmium (Cd) is a hazardous environmental pollutant that menaces human and animal health and induces serious adverse effects in various organs, particularly the liver and kidneys. Thus, the current study was designed to look into the possible mechanisms behind the ameliorative activities of Tamarindus indica (TM) and coenzyme Q10 (CoQ) combined therapy toward Cd-inflicted tissue injury. Male Wistar rats were categorized into seven groups: Control (received saline only); TM (50 mg/kg); CoQ (40 mg/kg); Cd (2 mg/kg); (Cd + TM); (Cd + CoQ); and (Cd + TM + CoQ). All the treatments were employed once daily via oral gavage for 28 consecutive days. The results revealed that Cd exposure considerably induced liver and kidney damage, evidenced by enhancement of liver and kidney function tests. In addition, Cd intoxication could provoke oxidative stress evidenced by markedly decreased glutathione (GSH) content and catalase (CAT) activity alongside a substantial increase in malondialdehyde (MDA) concentrations in the hepatic and renal tissues. Besides, disrupted protein and lipid metabolism were noticed. Unambiguously, TM or CoQ supplementation alleviated Cd-induced hepatorenal damage, which is most likely attributed to their antioxidant and anti-inflammatory contents. Interestingly, when TM and CoQ were given in combination, a better restoration of Cd-induced liver and kidney damage was noticed than was during their individual treatments.
Collapse
Affiliation(s)
- Amany Abdelnaby
- Department of Biotechnology, Animal Health Research Institute, Agricultural Research Center, Giza, Egypt
| | - Nabila Abdel-Aleem
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Benha University, Toukh, Egypt
| | - Ayman Mansour
- Department of Biotechnology, Animal Health Research Institute, Agricultural Research Center, Giza, Egypt
| | - Afaf Abdelkader
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Benha University, Benha, Egypt
- Center of Excellence in Screening of Environmental Contaminants (CESEC), Benha University, Toukh, Egypt
- *Correspondence: Afaf Abdelkader, ; Ahmed Abdeen,
| | - Amany N. Ibrahim
- Department of Pharmacology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Safwa M. Sorour
- Department of Pharmacology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Enas Elgendy
- Histology and Cell Biology Department, Faculty of Medicine, Benha University, Benha, Egypt
| | - Heba Bayoumi
- Histology and Cell Biology Department, Faculty of Medicine, Benha University, Benha, Egypt
| | - Shaymaa M. Abdelrahman
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Benha University, Benha, Egypt
| | - Samah F. Ibrahim
- Department of Clinical Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Ilhaam Alsaati
- Department of Basic Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Ahmed Abdeen
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Benha University, Toukh, Egypt
- Center of Excellence in Screening of Environmental Contaminants (CESEC), Benha University, Toukh, Egypt
- Department of Biochemistry, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
- *Correspondence: Afaf Abdelkader, ; Ahmed Abdeen,
| |
Collapse
|
13
|
Preclinical and Clinical Role of Coenzyme Q10 Supplementation in Various Pathological States. Drug Res (Stuttg) 2022; 72:367-371. [PMID: 35724675 DOI: 10.1055/a-1835-1738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Coenzyme Q10 (CoQ10) is an efficient antioxidant produced endogenously in a living organism. It acts as an important cofactor in the electron transport system of mitochondria and reported as a safe supplement in humans and animals with minimal adverse effect. CoQ10 is found naturally, as a trans configuration, chemical nomenclature of which is 2,3- dimethoxy-5- methyl-6-decaprenyle -1,4-benzoquinone. It is found in the body in two forms. In quinone form (oxidized form), it serves as an electron transporter that transfers the electrons in the electron transport chain between various complexes, and in ubiquinol form (reduced form), it serves as potent antioxidants by scavenging free radicals or by tocopherol regeneration in the living organism. Its primary roles include synthesis of adenosine triphosphate (ATP), stabilizes lipid membrane, antioxidant activity, cell growth stimulation, and cell death inhibition. CoQ10 has shown a variety of pharmacological and clinical effects including neuroprotective, hepatoprotective, anti-atherosclerotic, anticonvulsant, antidepressant, anti-inflammatory, antinociceptive, cardiovascular, antimicrobial, immunomodulatory, and various effects on the central nervous system. Present review has set about to bring updated information regarding to clinical and preclinical activities of CoQ10, which may be helpful to researchers to explore a new bioactive molecules for various therapeutic application.
Collapse
|
14
|
Inflammation and Nitro-oxidative Stress as Drivers of Endocannabinoid System Aberrations in Mood Disorders and Schizophrenia. Mol Neurobiol 2022; 59:3485-3503. [PMID: 35347586 DOI: 10.1007/s12035-022-02800-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 03/13/2022] [Indexed: 01/02/2023]
Abstract
The endocannabinoid system (ECS) is composed of the endocannabinoid ligands anandamide (AEA) and 2-arachidonoylgycerol (2-AG), their target cannabinoid receptors (CB1 and CB2) and the enzymes involved in their synthesis and metabolism (N-acyltransferase and fatty acid amide hydrolase (FAAH) in the case of AEA and diacylglycerol lipase (DAGL) and monoacylglycerol lipase (MAGL) in the case of 2-AG). The origins of ECS dysfunction in major neuropsychiatric disorders remain to be determined, and this paper explores the possibility that they may be associated with chronically increased nitro-oxidative stress and activated immune-inflammatory pathways, and it examines the mechanisms which might be involved. Inflammation and nitro-oxidative stress are associated with both increased CB1 expression, via increased activity of the NADPH oxidases NOX4 and NOX1, and increased CNR1 expression and DNA methylation; and CB2 upregulation via increased pro-inflammatory cytokine levels, binding of the transcription factor Nrf2 to an antioxidant response element in the CNR2 promoter region and the action of miR-139. CB1 and CB2 have antagonistic effects on redox signalling, which may result from a miRNA-enabled negative feedback loop. The effects of inflammation and oxidative stress are detailed in respect of AEA and 2-AG levels, via effects on calcium homeostasis and phospholipase A2 activity; on FAAH activity, via nitrosylation/nitration of functional cysteine and/or tyrosine residues; and on 2-AG activity via effects on MGLL expression and MAGL. Finally, based on these detailed molecular neurobiological mechanisms, it is suggested that cannabidiol and dimethyl fumarate may have therapeutic potential for major depressive disorder, bipolar disorder and schizophrenia.
Collapse
|
15
|
The Effect of Coenzyme Q10 on Liver Injury Induced by Valproic Acid and Its Antiepileptic Activity in Rats. Biomedicines 2022; 10:biomedicines10010168. [PMID: 35052847 PMCID: PMC8773341 DOI: 10.3390/biomedicines10010168] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/10/2022] [Accepted: 01/10/2022] [Indexed: 12/10/2022] Open
Abstract
Valproic acid (VPA) has toxic metabolites that can elevate oxidative stress markers, and the hepatotoxicity of VPA has been reported. Coenzyme Q10 (CoQ10) is one of the most widely used antioxidants. The effect of CoQ10 on epileptogenesis and VPA hepatotoxicity were examined. Rats were randomly divided into five groups: the control group received 0.5% methylcellulose by oral gavages daily and saline by intraperitoneal injection three times weekly. The PTZ group received 1% methylcellulose by gavages daily and 30 mg/kg PTZ by intraperitoneal injection three times weekly. The valproic acid group received 500 mg/kg valproic acid by gavage and 30 mg/kg PTZ, as above. The CoQ10 group received 200 mg/kg CoQ10 by gavages daily and 30 mg/kg PTZ, as above. The Valproic acid + CoQ10 group received valproic acid and CoQ10, as above. Results: CoQ10 exhibited anticonvulsant activity and potentiated the anticonvulsant effect of VPA. CoQ10 combined with VPA induced a more significant reduction in oxidative stress and improved the histopathological changes in the brain and liver compared to VPA treatment. In addition, CoQ10 reduced the level of toxic VPA metabolites. These findings suggest that the co-administration of CoQ10 with VPA in epilepsy might have therapeutic potential by increasing antiepileptic activity and reducing the hepatotoxicity of VPA.
Collapse
|
16
|
Chen S, Tang Y, Fang W, He T, Chen X, Zhang P. CoQ10 promotes resolution of necrosis and liver regeneration after acetaminophen-induced liver injury. Toxicol Sci 2021; 185:19-27. [PMID: 34668565 DOI: 10.1093/toxsci/kfab123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Coenzyme Q10 (CoQ10) which acts as an electron transporter in the mitochondrial respiratory chain has many beneficial effects on liver diseases. In our previous research, CoQ10 has been found to attenuate acetaminophen (APAP) induced acute liver injury (ALI). However, whether CoQ10 administration is still effective at the late stage of APAP overdose is still unknown. In this study, we aimed to test CoQ10 efficacy at the late stage of APAP overdose. C57BL/6J mice were intraperitoneally treated with APAP to induce liver injury. CoQ10 (5 mg/kg) was given to mice at 16 hours after APAP treatment. The results showed that while CoQ10 treatment at 16 hours post-APAP overdose had no effects on the expression of ROS generated genes or scavenged genes, it still significantly decreased necrosis of hepatocytes following APAP-induced ALI. Moreover, CoQ10 increased MerTK+ macrophages accumulation in the APAP-overdose liver and inhibition of MerTK signaling partly abrogated the protective role of CoQ10 treatment on the hepatic necrosis. CoQ10 treatment also significantly enhanced hepatocytes proliferation as shown in the increased BrdU incorporation in the APAP-intoxicated mice liver section. In addition, CoQ10 treatment increased hepatic PCNA and Cyclin D1 expression and promoted activation of the β-catenin signaling in APAP-overdose mice. To conclude, these data provide evidence that CoQ10 treatment is still effective at the late stage of APAP-induced ALI and promotes resolution of necrosis and liver regeneration following ALI.
Collapse
Affiliation(s)
- Shen Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, People's Republic of China
| | - Yi Tang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Wanjun Fang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Taiping He
- Department of Nutrition, School of Public Health, Guangdong Medical University, People's Republic of China
| | - Xu Chen
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Peiwen Zhang
- Department of Nutrition, School of Public Health, Guangdong Medical University, People's Republic of China
| |
Collapse
|
17
|
Influence of Systemic Administration of Coq10 Nanoparticles on Ischemia-Reperfusion Injury on Ovaries in Rat. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:2303417. [PMID: 34335800 PMCID: PMC8324357 DOI: 10.1155/2021/2303417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/30/2021] [Accepted: 07/15/2021] [Indexed: 11/17/2022]
Abstract
Using a rat ovary model, effects of COQ10 nanoparticles (NCOQ10) were studied on ischemia-reperfusion injury. In the present experimental study, following randomization of thirty healthy female Wistar rats ∼250 g, the animals were subjected to five experimental groups (n = 6): group SHAM : only laparotomy was performed, group IS: only a 3-hour ischemia was performed, group IS/REP: the procedure included a 3-hour ischemia followed by a 3-hour reperfusion, and 50 µL soybean oil (solvent of NCOQ10) was administered 30 min before cessation of reperfusion, group IS/NCOQ10: the procedure included a 3-hour ischemia only and 50 µL (0.3 mmol/lit/IP) of NCOQ10 30 min before cessation of ischemia, and group IS/REP/NCOQ10: the procedure included a 3-hour ischemia, a 3-hour reperfusion, and 20 µL (0.3 mmol/lit) of NCOQ10 30 min before cessation of ischemia. Significantly amended development of ischemia/reperfusion tissue injury was observed in animals treated with NCOQ10 compared to those of other groups (P=0.001). Mean values of biochemical indices were significantly higher than those observed for other groups (P=0.001). Significantly lower values of MDA were observed in IS/REP/NCOQ10 animals compared to those of other groups (P=0.001).Where ovarian tissue is exposed to ischemia, intraperitoneal administration of NCOQ10 could bear clinical benefits in diminishing ischemia-reperfusion injury.
Collapse
|
18
|
Onaolapo OJ, Omotoso SA, Olofinnade AT, Onaolapo AY. Anti-inflammatory, anti-oxidant and anti-lipaemic effects of daily dietary coenzyme-Q10 supplement in a mouse model of metabolic syndrome. Antiinflamm Antiallergy Agents Med Chem 2021; 20:380-388. [PMID: 33906592 DOI: 10.2174/1871523020666210427111328] [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] [Received: 01/20/2021] [Revised: 03/16/2021] [Accepted: 03/29/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The dietary model of metabolic syndrome has continued to aid our understanding of its pathogenesis and possible management interventions. However, despite progress in research, therapy continues to be challenging in humans; hence, the search for newer treatment and prevention options. OBJECTIVE To evaluate the impact of dietary CQ10 supplementation on metabolic, oxidative and inflammatory markers in a diet-induced mouse model of metabolic syndrome. METHODS Mouse groups were fed standard diet (SD), high-fat high-sugar (HFHS) diet, and SD or HFHS diet (with incorporated CQ10) at 60 and 120 mg/kg of feed respectively. At the completion of the study (8 weeks), blood glucose levels, superoxide dismutase (SOD) activity, plasma insulin, leptin, adiponectin, TNF-α, IL-10, serum lipid profile, and lipid peroxidation (LPO) levels were assessed. The liver was either homogenised for the assessment of antioxidant status or processed for general histology. RESULTS Dietary CQ10 mitigated HFHS diet-induced weight gain, decreased glucose, insulin and leptin levels; and increased adiponectin levels in mice. Coenzyme-Q10 improved the antioxidant status of the liver and blood in HFHS diet fed mice, while also decreasing lipid peroxidation. Lipid profile improved, level of TNF-α decreased and IL-10 increased following CQ10 diet. A mitigation of HFHS diet-induced alteration in liver morphology was also observed with CQ10. CONCLUSION Dietary CQ10 supplementation mitigates HFHS diet-induced changes in mice possibly through its anti-oxidant, anti-lipaemic and anti-inflammatory potential.
Collapse
Affiliation(s)
- Olakunle J Onaolapo
- Behavioural Neuroscience/Neuropharmacology Unit, Department of Pharmacology, Ladoke Akintola University of Technology, Ogbomosho, Oyo State. Nigeria
| | - Sarafa A Omotoso
- Behavioural Neuroscience/Neuropharmacology Unit, Department of Pharmacology, Ladoke Akintola University of Technology, Ogbomosho, Oyo State. Nigeria
| | - Anthony T Olofinnade
- Behavioural Neuroscience/Neuropharmacology Unit, Department of Pharmacology, Ladoke Akintola University of Technology, Ogbomosho, Oyo State. Nigeria
| | - Adejoke Y Onaolapo
- Behavioural Neuroscience/Neurobiology Unit, Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho, Oyo State. Nigeria
| |
Collapse
|
19
|
Abiri B, Vafa M. Impact of coenzyme Q10 on inflammatory biomarkers and its role in future therapeutic strategies. Clin Nutr ESPEN 2021; 43:25-30. [PMID: 34024523 DOI: 10.1016/j.clnesp.2021.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/12/2021] [Accepted: 04/07/2021] [Indexed: 11/15/2022]
Abstract
Coenzyme Q (CoQ) is an important component of the mitochondrial electron transport chain. The finding that multiple chronic diseases show lower levels of CoQ10 has led to the possibility that CoQ10 supplementation could be an effective approach to ameliorate or prevent disease progression. In this review, we discuss the state of the art regarding the role of CoQ10 in health and disease and describe the latest clinical studies which have tested the effects of CoQ10 supplementation in inflammatory diseases. The results of these studies indicate that individuals suffering from inflammation-related diseases show improvement under the CoQ10 supplementation protocol. However, these results have been inconsistent, leading to the need for additional studies at the preclinical and clinical levels, involving a greater number of subjects and different treatment regimes.
Collapse
Affiliation(s)
- Behnaz Abiri
- Nutritional Sciences, Department of Nutrition, Faculty of Paramedicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammadreza Vafa
- Nutritional Sciences, Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
20
|
Oyovwi MO, Nwangwa EK, Ben-Azu B, Edesiri TP, Emojevwe V, Igweh JC. Taurine and coenzyme Q10 synergistically prevent and reverse chlorpromazine-induced psycho-neuroendocrine changes and cataleptic behavior in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2021; 394:717-734. [PMID: 33146779 DOI: 10.1007/s00210-020-02003-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 10/15/2020] [Indexed: 12/21/2022]
Abstract
Over the years, mounting evidences have suggested a strong association between chronic chlorpromazine therapy, a popular first-generation antipsychotic drug, and psycho-neuroendocrine changes. In this study, we aim to examine whether treatment with taurine and coenzyme Q10 (COQ-10), compounds with steroidogenic-gonadotropin hormone-enhancing properties, can attenuate the negative impacts of chlorpromazine on steroidogenic, gonadotropin, thyroid and HPA-axis hormones, dopamine levels, catalepsy behavior and neuronal cells of the hypothalamus and pituitary gland in the preventive and reversal treatments in male Wister rats. In the drug treatment alone or preventive protocol, rats received oral administration of saline (10 mL/kg), taurine (150 mg/kg/day), COQ-10 (10 mg/kg/day), or both (taurine + COQ-10/day) alone for 56 consecutive days, or in combination with oral chlorpromazine (30 mg/kg/day) treatment from days 29 to 56. In the reversal protocol, the animals received chlorpromazine or saline for 56 days prior to taurine, COQ-10, or the combination from days 29 to 56. Thereafter, serum prolactin, steroidogenic (testosterone, estrogen, progesterone), gonadotropin (luteinizing hormone, LH, follicle-stimulating hormone, FSH), thyroid (thyrotropin-stimulating hormone, tetraiodothyronine, triiodothyronine) hormones, corticosterone, brain dopamine levels and cataleptic behavior were investigated. The histopathological features of the hypothalamus and pituitary gland were also evaluated. Taurine, COQ-10, or their combination prevented and reversed chlorpromazine-induced hyperprolactinemia, decrease in FSH, LH, testosterone, progesterone and dopamine concentrations, as well as the increase in estrogen levels. Taurine and COQ-10 reduced the changes in thyroid hormones, corticosterone release, histological distortions of the hypothalamus and the pituitary gland of chlorpromazine-treated rats. Taurine and COQ-10 attenuated chlorpromazine-induced catalepsy. The study showed that taurine and COQ-10 prevented and reversed chlorpromazine-induced changes in reproductive, thyroid hormones, dopamine level, corticosterone release, neurodegenerations, and cataleptic behavior in rats.
Collapse
Affiliation(s)
- Mega O Oyovwi
- Department of Human Physiology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Delta State, Nigeria
- Department of Basic Medical Science, Achievers University, Owo, Ondo State, Nigeria
| | - Eze K Nwangwa
- Department of Human Physiology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Benneth Ben-Azu
- Department of Pharmacology, Faculty of Basic Medical Science, PAMO University of Medical Sciences, Port Harcourt, Rivers State, Nigeria.
| | - Tesi P Edesiri
- Department of Science Laboratory Technology, Delta State Polytechnic, Ogwashi-Uku, Delta State, Nigeria
| | - Victor Emojevwe
- Department of Physiology, Faculty of Baic Medical Sciences, University of Medical Sciences, Ondo, Ondo State, Nigeria
| | - John C Igweh
- Department of Human Physiology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Delta State, Nigeria
| |
Collapse
|
21
|
Mohsen AM, Younis MM, Salama A, Darwish AB. Cubosomes as a Potential Oral Drug Delivery System for Enhancing the Hepatoprotective Effect of Coenzyme Q10. J Pharm Sci 2021; 110:2677-2686. [PMID: 33600809 DOI: 10.1016/j.xphs.2021.02.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/05/2021] [Accepted: 02/05/2021] [Indexed: 10/22/2022]
Abstract
Coenzyme Q10 (CoQ10) acts as an antioxidant that protects the cells by preventing lipid peroxidation. Owing to its low solubility, CoQ10 has shown poor delivery properties and poor bioavailability. The aim of this study is to develop CoQ10 loaded cubosomes in order to enhance its oral delivery and hepatoprotective activity. Cubosomes are cubic nanostructured systems resulting from the colloidal dispersion of cubic liquid crystalline structure in water. CoQ10 loaded cubosomes were prepared using poloxamer 407 and glyceryl monooleate at three weight ratios (1:2.5, 1:5 and 1:7.5) and were further characterized. They were investigated for their hepatoprotective effect in thioacetamide (TAA) induced hepatotoxicity in Wistar rats. The developed CoQ10 cubosomes exhibited moderate to high entrapment efficiency percentages (44.69-75.96%), nanometric dimensions (132.4-223.2 nm), and negatively charged zeta potential values (<-21.3). In-vitro release profiles showed a sustained release of CoQ10 from the developed cubosomes up to 48 h. In-vivo study revealed an improved hepatoprotective effect of CoQ10 cubosomes via reducing liver enzymes, nitric oxide and malondialdehyde as well as elevating phosphoinositide 3-kinase, catalase and glutathione peroxidase, compared to plain drug. These results were in good agreement with histopathological investigations. Consequently, the developed cubosomes showed a potential effect in enhancing the hepatoprotective activity of CoQ10.
Collapse
Affiliation(s)
- Amira Mohamed Mohsen
- Pharmaceutical Technology Department, National Research Centre, Dokki, Cairo, Egypt.
| | | | - Abeer Salama
- Pharmacology Department, National Research Centre, Dokki, Cairo, Egypt
| | - Asmaa Badawy Darwish
- Pharmaceutical Technology Department, National Research Centre, Dokki, Cairo, Egypt
| |
Collapse
|
22
|
Zhang P, Chen S, Tang H, Fang W, Chen K, Chen X. CoQ10 protects against acetaminophen-induced liver injury by enhancing mitophagy. Toxicol Appl Pharmacol 2020; 410:115355. [PMID: 33271250 DOI: 10.1016/j.taap.2020.115355] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 11/11/2020] [Accepted: 11/26/2020] [Indexed: 12/24/2022]
Abstract
Coenzyme Q10 (CoQ10), which is a key cofactor of the electron transport chain in the mitochondria has shown many beneficial effects on liver diseases. However, the mechanisms of CoQ10 protective role on the acetaminophen (APAP)-induced liver injury are elusive and unclear. In this study, we further investigated the CoQ10 therapeutic effects on APAP-overdose liver injury. C57BL/6 J mice were intraperitoneally treated with APAP to induce liver injury. CoQ10 (5 mg/kg) was given to mice at 1.5 h after APAP treatment. The results showed that hepatic CoQ10 levels were decreased during the APAP-induced hepatotoxicity and preceded serum ALT elevation. Treatment of CoQ10 significantly improved the liver injury induced by APAP. Moreover, CoQ10 treatment decreased the ROS levels and promoted the antioxidative related gene expression in APAP overdose mice. Importantly, results showed that even though CoQ10 had no effects on the mtDNA copy number and the expression of genes related to mitochondrial biogenesis, it significantly improved the mitochondrial complex I and V activities and promoted the mitophagy in APAP-overdose mice. To further authenticate mitophagy role in CoQ10-mediated improved liver injury in vivo, we administrated APAP-overdose mice with chloroquine 1 h prior to APAP treatment and found that chloroquine treatment functionally abrogated the CoQ10 protective role on APAP overdose mice. To conclude, this study provides evidence that CoQ10 activates mitophagy to protect against APAP-induced liver injury. Therefore, CoQ10 may represent a novel therapeutic option for the prevention and treatment of drug-induced liver injury.
Collapse
Affiliation(s)
- Peiwen Zhang
- Department of Nutrition, School of Public Health, Guangdong Medical University, People's Republic of China; School of Public Health, Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, People's Republic of China
| | - Shen Chen
- Department of Nutrition, School of Public Health, Guangdong Medical University, People's Republic of China; Department of Toxicology, School of Public Health, Sun Yat-sen University, People's Republic of China
| | - Huanwen Tang
- School of Public Health, Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, People's Republic of China
| | - Wanjun Fang
- Department of Clinical Nutrition, Ningbo Women and Children's Hospital, Ningbo, People's Republic of China
| | - Ke Chen
- Department of Clinical Nutrition, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, People's Republic of China; Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Xu Chen
- Department of Nutrition, School of Public Health, Guangdong Medical University, People's Republic of China; Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, People's Republic of China.
| |
Collapse
|
23
|
The interplay between oxidative stress and bioenergetic failure in neuropsychiatric illnesses: can we explain it and can we treat it? Mol Biol Rep 2020; 47:5587-5620. [PMID: 32564227 DOI: 10.1007/s11033-020-05590-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 06/12/2020] [Indexed: 12/12/2022]
Abstract
Nitro-oxidative stress and lowered antioxidant defences play a key role in neuropsychiatric disorders such as major depression, bipolar disorder and schizophrenia. The first part of this paper details mitochondrial antioxidant mechanisms and their importance in reactive oxygen species (ROS) detoxification, including details of NO networks, the roles of H2O2 and the thioredoxin/peroxiredoxin system, and the relationship between mitochondrial respiration and NADPH production. The second part highlights and identifies the causes of the multiple pathological sequelae arising from self-amplifying increases in mitochondrial ROS production and bioenergetic failure. Particular attention is paid to NAD+ depletion as a core cause of pathology; detrimental effects of raised ROS and reactive nitrogen species on ATP and NADPH generation; detrimental effects of oxidative and nitrosative stress on the glutathione and thioredoxin systems; and the NAD+-induced signalling cascade, including the roles of SIRT1, SIRT3, PGC-1α, the FOXO family of transcription factors, Nrf1 and Nrf2. The third part discusses proposed therapeutic interventions aimed at mitigating such pathology, including the use of the NAD+ precursors nicotinamide mononucleotide and nicotinamide riboside, both of which rapidly elevate levels of NAD+ in the brain and periphery following oral administration; coenzyme Q10 which, when given with the aim of improving mitochondrial function and reducing nitro-oxidative stress in the brain, may be administered via the use of mitoquinone, which is in essence ubiquinone with an attached triphenylphosphonium cation; and N-acetylcysteine, which is associated with improved mitochondrial function in the brain and produces significant decreases in oxidative and nitrosative stress in a dose-dependent manner.
Collapse
|
24
|
Impact of Coenzyme Q10 Administration on Lead Acetate-Induced Testicular Damage in Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4981386. [PMID: 32566085 PMCID: PMC7290879 DOI: 10.1155/2020/4981386] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/28/2020] [Accepted: 05/12/2020] [Indexed: 12/30/2022]
Abstract
Exposure to lead (Pb) causes multiorgan dysfunction including reproductive impairments. Here, we examined the protective effects of coenzyme Q10 (CoQ10) administration on testicular injury induced by lead acetate (PbAc) exposure in rats. This study employed four experimental groups (n = 7) that underwent seven days of treatment as follows: control group intraperitoneally (i.p.) treated with 0.1 ml of 0.9% NaCl containing 1% Tween 80 (v : v), CoQ10 group that was i.p. injected with 10 mg/kg CoQ10, PbAc group that was i.p. treated with PbAc (20 mg/kg), and PbAc+CoQ10 group that was i.p. injected with CoQ10 2 h after PbAc. PbAc injection resulted in increasing residual Pb levels in the testis and reducing testosterone, luteinizing hormone, and follicle-stimulating hormone levels. Additionally, PbAc exposure resulted in significant oxidative damage to the tissues on the testes. PbAc raised the levels of prooxidants (malondialdehyde and nitric oxide) and reduced the amount of endogenous antioxidative proteins (glutathione and its derivative enzymes, catalase, and superoxide dismutase) available in the cell. Moreover, PbAc induced the inflammatory response as evidenced by the upregulation of inflammatory mediators (tumor necrosis factor-alpha and interleukin-1 beta). Further, PbAc treatment induced apoptosis in the testicular cells, as indicated by an increase in Bax and caspase 3 expression, and reduced Bcl2 expression. CoQ10 supplementation improved testicular function by inhibiting Pb accumulation, oxidative stress, inflammation, cell death, and histopathological changes following PbAc exposure. Our findings suggest that CoQ10 can act as a natural therapeutic agent to protect against the reproductive impairments associated with PbAc exposure.
Collapse
|
25
|
Kim HN, Jeon DG, Lim Y, Jang IS. The effects of coenzyme Q 10 supplement on blood lipid indices and hepatic antioxidant defense system in SD rats fed a high cholesterol diet. Lab Anim Res 2020; 35:13. [PMID: 32257901 PMCID: PMC7081576 DOI: 10.1186/s42826-019-0013-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/24/2019] [Indexed: 12/19/2022] Open
Abstract
A total of 24 SD rats were allotted to four treatment groups such as the control (CON), 1% of cholesterol diet (CHO), 0.5% of coenzyme Q10 (COQ) and 1% of cholesterol plus 0.5% of coenzyme Q10 (CHCQ) groups to determine the effects of coenzyme Q10 (CoQ10) on the antioxidant defense system in rats. The body weight, weight gain, liver weight and abdominal fat pads were unaffected by 0.5% of CoQ10 supplement in the rats. The level of triglyceride and HDL-cholesterol levels in the blood was significantly increased (p < 0.05) by the 1% of cholesterol supplement (CHO), whereas 0.5% of CoQ10 supplement (COQ) did not alter these blood lipid indices. In the mRNA expression, there was a significant effect (P < 0.05) of the CoQ10 supplement on the mRNA expression of superoxide dismutase (SOD), although the mRNA expression of glutathione peroxidase (GPX) and glutathione S-transferase (GST) was unaffected by cholesterol or the CoQ10 supplement. Similar to mRNA expression of SOD, its activity was also significantly increased (P < 0.05) by CoQ10, but not by the cholesterol supplement effect. The activities hepatic GPX and GST were unaffected by CoQ10 and cholesterol supplements in rats. Lipid peroxidation in the CHO group resulted in a significant (p < 0.05) increase compared with that in the other groups, indicating that the CoQ10 supplement to 1% of cholesterol-fed rats alleviated the production of lipid peroxidation in the liver. In conclusion, 0.5% of the CoQ10 supplement resulted in positive effects on the hepatic antioxidant defense system without affecting blood lipid indices in 1% of cholesterol fed rats.
Collapse
Affiliation(s)
- Ha-Na Kim
- 1Department of Animal Science and Biotechnology, and the Regional Animal Research Center, Gyeongnam National University of Science and Technology, Chilam-Dong 150, Jinju, Gyeongnam 52725 Korea
| | - Dong-Gyung Jeon
- 1Department of Animal Science and Biotechnology, and the Regional Animal Research Center, Gyeongnam National University of Science and Technology, Chilam-Dong 150, Jinju, Gyeongnam 52725 Korea
| | - Yong Lim
- 2Department of Clinical Laboratory Science, Dong-Eui Univerisity, Busan, 47340 Korea
| | - In-Surk Jang
- 1Department of Animal Science and Biotechnology, and the Regional Animal Research Center, Gyeongnam National University of Science and Technology, Chilam-Dong 150, Jinju, Gyeongnam 52725 Korea
| |
Collapse
|
26
|
Elshazly SM, Alsemeh AE, Ahmad EAA, Rezq S. CoQ10 exerts hepatoprotective effect in fructose-induced fatty liver model in rats. Pharmacol Rep 2020; 72:922-934. [PMID: 32157594 DOI: 10.1007/s43440-020-00075-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 11/02/2019] [Accepted: 12/30/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Excess dietary sugar is associated with deleterious metabolic effects, liver injury, and coenzyme-Q10 (CoQ10) deficiency. This study investigates the ability of CoQ10 to protect against fructose-induced hepatic damage. METHODS Rats were fed tap water or 30% fructose for 12 weeks with or without CoQ10 (10 mg/kg, po). An additional group of rats were allowed to feed on either water or 30% fructose for 12 weeks, followed by four weeks of treatment with either the vehicle or CoQ10. RESULTS Fructose-fed rats showed lower CoQ10 levels, increased systolic pressure, increased body weight, higher liquid consumption, decreased food intake and hyperglycemia. Fructose-fed rats also showed deteriorated serum and liver lipid profiles, impaired liver function tests and oxidative status, and lower expression of adiponectin receptor 1 and 2 along with higher GLUT-2 levels. Furthermore, following fructose treatment, tyrosine kinase-PI3K pathway was inhibited. Additionally, there was an increase in the levels of apoptotic markers and serum visfatin and a decrease in the levels of adiponectin and soluble receptor of the advanced glycated end product. Consequently, several histopathological changes were detected in the liver. Concurrent or three months post-exposure administration of CoQ10 in fructose rats significantly reversed or attenuated all the measured parameters and hepato-cytoarchitecture alterations. CONCLUSION This study suggests CoQ10 supplement as a possible prophylaxis or treatment candidate for fructose-induced liver injury.
Collapse
Affiliation(s)
- Shimaa M Elshazly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Amira E Alsemeh
- Department of Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Enssaf A A Ahmad
- Department of Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Samar Rezq
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt. .,Department of Cell and Molecular Biology, UMMC, 2500 N state St., Jackson, MS, 39216, USA.
| |
Collapse
|
27
|
Mantle D, Hargreaves IP. Coenzyme Q10 supplementation in non-alcoholic fatty liver disease: an overview. GASTROINTESTINAL NURSING 2020; 18:22-27. [DOI: 10.12968/gasn.2020.18.2.22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common liver disorder in the UK, for which little effective conventional treatment is available. Mitochondrial dysfunction, oxidative stress and inflammation have been implicated in the pathogenesis of NAFLD. This article focuses on the role of the vitamin-like substance coenzyme Q10 (CoQ10) in NAFLD, since CoQ10 plays a key role in mitochondrial function, as well as having antioxidant and anti-inflammatory action. CoQ10 levels are depleted in NAFLD, and studies in animal models and human subjects have indicated that supplementation with CoQ10 can significantly reduce oxidative stress and the inflammation characteristic of NAFLD. In addition, NAFLD patients are at increased risk of developing heart failure, and supplementary CoQ10 may help to reduce this risk. Supplementary CoQ10 is generally well tolerated, with no significant adverse effects reported in long-term use.
Collapse
|
28
|
Neag MA, Catinean A, Muntean DM, Pop MR, Bocsan CI, Botan EC, Buzoianu AD. Probiotic Bacillus Spores Protect Against Acetaminophen Induced Acute Liver Injury in Rats. Nutrients 2020; 12:nu12030632. [PMID: 32120994 PMCID: PMC7146158 DOI: 10.3390/nu12030632] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 02/18/2020] [Accepted: 02/24/2020] [Indexed: 12/17/2022] Open
Abstract
Acetaminophen (APAP) is one of the most used analgesics and antipyretic agents in the world. Intoxication with APAP is the main cause of acute liver toxicity in both the US and Europe. Spore-forming probiotic bacteria have the ability to resist harsh gastric and intestinal conditions. The aim of this study was to investigate the possible protective effect of Bacillus (B) species (sp) spores (B. licheniformis, B. indicus, B. subtilis, B. clausii, B. coagulans) against hepatotoxicity induced by APAP in rats. A total of 35 rats were randomly divided into seven groups: group I served as control; group II received silymarin; group III received MegaSporeBioticTM (MSB); group IV received APAP and served as the model of hepatotoxicity; group V received APAP and silymarin; group VI received APAP and MSB; group VII received APAP, silymarin and MSB. The livers for histopathological examination and blood samples were collected on the last day of the experiment. We determined aspartate aminotransferase (AST), alanine aminotransferase (ALT) and total antioxidant capacity (TAC) levels and zonula occludens (ZO-1), tumor necrosis factor α (TNF-α) and interleukin 1β (IL-1β) expression. APAP overdose increased AST and ALT. It slowly decreased TAC compared to the control group, but pretreatment with silymarin and MSB increased TAC levels. Elevated plasma concentrations were identified for ZO-1 in groups treated with APAP overdose compared with those without APAP or receiving APAP in combination with silymarin, MSB or both. The changes were positively correlated with the levels of other proinflammatory cytokines (TNF-α, IL-1β). In addition, histopathological hepatic injury was improved by preadministration of MSB or silymarin versus the disease model group. Bacillus sp spores had a protective effect on acute hepatic injury induced by APAP. Pretreatment with MSB resulted in a significant reduction in serum AST, ALT, TNF-α, IL-1β, ZO-1, TAC and also hepatocyte necrosis, similar to the well-known hepatoprotective agent—silymarin.
Collapse
Affiliation(s)
- Maria Adriana Neag
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400337, Romania; (M.A.N.); (M.R.P.); (C.I.B.); (A.D.B.)
| | - Adrian Catinean
- Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400006, Romania
- Correspondence: ; Tel.: +40-752122466
| | - Dana Maria Muntean
- Department of Pharmaceutical Technology and Biopharmaceutics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400010, Romania;
| | - Maria Raluca Pop
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400337, Romania; (M.A.N.); (M.R.P.); (C.I.B.); (A.D.B.)
| | - Corina Ioana Bocsan
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400337, Romania; (M.A.N.); (M.R.P.); (C.I.B.); (A.D.B.)
| | | | - Anca Dana Buzoianu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400337, Romania; (M.A.N.); (M.R.P.); (C.I.B.); (A.D.B.)
| |
Collapse
|
29
|
AL-Megrin WA, Soliman D, Kassab RB, Metwally DM, Ahmed E. Abdel Moneim, El-Khadragy MF. Coenzyme Q10 Activates the Antioxidant Machinery and Inhibits the Inflammatory and Apoptotic Cascades Against Lead Acetate-Induced Renal Injury in Rats. Front Physiol 2020; 11:64. [PMID: 32116774 PMCID: PMC7020615 DOI: 10.3389/fphys.2020.00064] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 01/21/2020] [Indexed: 12/29/2022] Open
Abstract
The kidney is among the metabolic organs most susceptible to injury, particularly following exposure to xenobiotics and heavy metals. We aimed to explore the potential protective impacts of coenzyme Q10 (CoQ10) on lead acetate (PbAc)-induced nephrotoxicity in rats. Four experimental groups (n = 7) were applied as follows: control group, CoQ10 alone (10 mg/kg), PbAc alone (20 mg/kg), and PbAc with CoQ10. Exposure to PbAc led to the accumulation of Pb in the kidney and increased urea and creatinine serum levels. The deposition of Pb coupled with the elevation of malondialdehyde and nitrate/nitrite levels along with the upregulation of inducible nitric oxide synthase. Additionally, upon PbAc poisoning, glutathione content and the antioxidant enzymes were depleted along with the downregulation of Nrf2 and HO-1 expression. Moreover, PbAc injection increased the protein and mRNA levels of pro-inflammatory cytokines namely, tumor necrosis factor-alpha and interleukin-1 beta, while decreased the levels of interleukin-10, an anti-inflammatory cytokine, in the kidney. Furthermore, exposure to PbAc correlated with increased levels of pro-apoptotic markers, Bax and caspase-3, and reduced levels of the anti-apoptotic marker Bcl-2. The administration of CoQ10 alleviated the molecular, biochemical and histological changes following PbAc intoxication. Thus, CoQ10 reduces the deleterious cellular side effects of PbAc exposure due to its antioxidant, anti-inflammatory and anti-apoptotic effects.
Collapse
Affiliation(s)
- Wafa A. AL-Megrin
- Department of Biology, Faculty of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Doaa Soliman
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| | - Rami B. Kassab
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| | - Dina M. Metwally
- Department of Parasitology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
- Department of Zoology, Faculty of Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed E. Abdel Moneim
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| | - Manal F. El-Khadragy
- Department of Biology, Faculty of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| |
Collapse
|
30
|
Morris G, Puri BK, Walker AJ, Berk M, Walder K, Bortolasci CC, Marx W, Carvalho AF, Maes M. The compensatory antioxidant response system with a focus on neuroprogressive disorders. Prog Neuropsychopharmacol Biol Psychiatry 2019; 95:109708. [PMID: 31351160 DOI: 10.1016/j.pnpbp.2019.109708] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/16/2019] [Accepted: 07/22/2019] [Indexed: 02/07/2023]
Abstract
Major antioxidant responses to increased levels of inflammatory, oxidative and nitrosative stress (ONS) are detailed. In response to increasing levels of nitric oxide, S-nitrosylation of cysteine thiol groups leads to post-transcriptional modification of many cellular proteins and thereby regulates their activity and allows cellular adaptation to increased levels of ONS. S-nitrosylation inhibits the function of nuclear factor kappa-light-chain-enhancer of activated B cells, toll-like receptor-mediated signalling and the activity of several mitogen-activated protein kinases, while activating nuclear translocation of nuclear factor (erythroid-derived 2)-like 2 (Nrf2 or NFE2L2); in turn, the redox-regulated activation of Nrf2 leads to increased levels and/or activity of key enzymes and transporter systems involved in the glutathione system. The Nrf2/Kelch-like ECH-associated protein-1 axis is associated with upregulation of NAD(P)H:quinone oxidoreductase 1, which in turn has anti-inflammatory effects. Increased Nrf2 transcriptional activity also leads to activation of haem oxygenase-1, which is associated with upregulation of bilirubin, biliverdin and biliverdin reductase as well as increased carbon monoxide signalling, anti-inflammatory and antioxidant activity. Associated transcriptional responses, which may be mediated by retrograde signalling owing to elevated hydrogen peroxide, include the unfolded protein response (UPR), mitohormesis and the mitochondrial UPR; the UPR also results from increasing levels of mitochondrial and cytosolic reactive oxygen species and reactive nitrogen species leading to nitrosylation, glutathionylation, oxidation and nitration of crucial cysteine and tyrosine causing protein misfolding and the development of endoplasmic reticulum stress. It is shown how these mechanisms co-operate in forming a co-ordinated rapid and prolonged compensatory antioxidant response system.
Collapse
Affiliation(s)
- Gerwyn Morris
- IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Basant K Puri
- Department of Medicine, Hammersmith Hospital, Imperial College London, London, United Kingdom
| | - Adam J Walker
- IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Michael Berk
- IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Deakin University, Geelong, VIC, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry, The Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Ken Walder
- CMMR Strategic Research Centre, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Chiara C Bortolasci
- CMMR Strategic Research Centre, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Wolfgang Marx
- IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Andre F Carvalho
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada.
| | - Michael Maes
- IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| |
Collapse
|
31
|
Coenzyme Q10 Cardioprotective Effects Against Doxorubicin-Induced Cardiotoxicity in Wistar Rat. Cardiovasc Toxicol 2019; 20:222-234. [DOI: 10.1007/s12012-019-09547-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
32
|
The Neuroprotective Role of Coenzyme Q10 Against Lead Acetate-Induced Neurotoxicity Is Mediated by Antioxidant, Anti-Inflammatory and Anti-Apoptotic Activities. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16162895. [PMID: 31412628 PMCID: PMC6720293 DOI: 10.3390/ijerph16162895] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/16/2019] [Accepted: 07/18/2019] [Indexed: 12/15/2022]
Abstract
Heavy metal exposure, in lead (Pb) particularly, is associated with severe neuronal impairment though oxidative stress mediated by reactive oxygen species, and antioxidants may be used to abolish these adverse effects. This study investigated the potential neuroprotective role of coenzyme Q10 (CoQ10) against lead acetate (PbAc)-induced neurotoxicity. Twenty-eight male Wistar albino rats were divided into four equal groups (n = 7) and treated as follows: the control group was injected with physiological saline (0.9% NaCl); the CoQ10 group was injected with CoQ10 (10 mg/kg); PbAc group was injected with PbAc (20 mg/kg); PbAc + CoQ10 group was injected first with PbAc, and after 1 h with CoQ10. All groups were injected intraperitoneally for seven days. PbAc significantly increased cortical lipid peroxidation, nitrate/nitrite levels, and inducible nitric oxide synthase expression, and decreased glutathione content, superoxide dismutase, catalase, glutathione peroxidase, glutathione reductase activity and mRNA expression, as well as nuclear factor erythroid 2–related factor 2 (Nrf2) and homoxygenase-1 (HO-1) expression. PbAc also promoted the secretion of interleukin-1ß and tumor necrosis factor-α, inhibited interleukin-10 production, triggered the activation of pro-apoptotic proteins, and suppressed anti-apoptotic proteins. Additionally, PbAc increased the cortical levels of serotonin, dopamine, norepinephrine, GABA, and glutamate, and decreased the level of ATP. However, treatment with CoQ10 rescued cortical neurons from PbAc-induced neurotoxicity by restoring the balance between oxidants and antioxidants, activating the Nrf2/HO-1 pathway, suppressing inflammation, inhibiting the apoptotic cascade, and modulating cortical neurotransmission and energy metabolism. Altogether, our findings indicate that CoQ10 has beneficial effects against PbAc-induced neuronal damage through its antioxidant, anti-inflammatory, anti-apoptotic, and neuromodulatory activities.
Collapse
|
33
|
Ahmed OM, Fahim HI, Ahmed HY, Al-Muzafar HM, Ahmed RR, Amin KA, El-Nahass ES, Abdelazeem WH. The Preventive Effects and the Mechanisms of Action of Navel Orange Peel Hydroethanolic Extract, Naringin, and Naringenin in N-Acetyl-p-aminophenol-Induced Liver Injury in Wistar Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2745352. [PMID: 31049130 PMCID: PMC6458942 DOI: 10.1155/2019/2745352] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/24/2018] [Indexed: 01/14/2023]
Abstract
N-Acetyl-p-aminophenol (APAP) or acetaminophen is the most common drug ingredient worldwide. It is found in more than 600 different over-the-counter and prescription medicines. Its long-term and overdose use is highly toxic and may result in liver injury. Thus, this study was designed to assess the preventive effects and to suggest the mechanisms of action of the navel orange peel hydroethanolic extract, naringin, and naringenin in APAP-induced hepatotoxicity in male Wistar rats. APAP was administered to male Wistar rats at a dose level of 0.5 g/kg body weight (b.w.) by oral gavage every other day for 4 weeks. APAP-administered rats were treated with the navel orange peel hydroethanolic extract (50 mg/kg b.w.), naringin (20 mg/kg b.w.), and naringenin (20 mg/kg b.w.) by oral gavage every other day during the same period of APAP administration. The treatments of APAP-administered rats with the peel extract, naringin, and naringenin produced a significant decrease in the elevated serum AST, ALT, ALP, LDH, and GGT activities as well as total bilirubin and TNF-α levels while they induced a significant increase in the lowered serum albumin and IL-4 levels. The treatments also resulted in a significant decrease in the elevated liver lipid peroxidation and enhanced the liver GSH content and SOD, GST, and GPx activities as compared with APAP-administered control; the peel extract was the most potent in improving the liver LPO, GSH content, and GPx activity. In addition, the three treatments significantly downregulated the elevated hepatic proapoptotic mediators p53, Bax, and caspase-3 and significantly upregulated the suppressed antiapoptotic protein, Bcl-2, in APAP-administered rats. In association, the treatments markedly amended the APAP-induced liver histopathological deteriorations that include hepatocyte steatosis, cytoplasmic vacuolization, hydropic degeneration, and necrosis together with mononuclear leucocytic and fibroblastic inflammatory cells' infiltration. In conclusion, the navel orange peel hydroethanolic extract, naringin, and naringenin may exert their hepatopreventive effects in APAP-administered rats via enhancement of the antioxidant defense system and suppression of inflammation and apoptosis.
Collapse
Affiliation(s)
- Osama M. Ahmed
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Hanaa I. Fahim
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Heba Y. Ahmed
- Rodents Division, Department of Harmful Animals, Plant Protection Research Institute, Agriculture Research Center, Egypt
| | - Hessah Mohammed Al-Muzafar
- Chemistry Department, College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Rasha R. Ahmed
- Cell Biology, Histology and Genetics Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Kamal Adel Amin
- Chemistry Department, College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - El-Shaymaa El-Nahass
- Department of Pathology, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Walaa H. Abdelazeem
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
34
|
Yuksel TN, Yayla M, Halici Z, Cadirci E, Polat B, Kose D. Protective effect of 5-HT7 receptor activation against glutamate-induced neurotoxicity in human neuroblastoma SH-SY5Y cells via antioxidative and antiapoptotic pathways. Neurotoxicol Teratol 2019; 72:22-28. [PMID: 30685503 DOI: 10.1016/j.ntt.2019.01.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 12/23/2018] [Accepted: 01/17/2019] [Indexed: 12/29/2022]
Abstract
Serotonin exerts anti-inflammatory, antioxidant and antiapoptotic effects through 5-HT7 receptors. The present study determined the role of 5-HT7 receptors in glutamate-induced neurotoxicity by using human SH-SY5Y neuroblastoma cells. The cells were pretreated with different concentrations of 5-HT7 receptor agonist LP44 and antagonist SB269970 for 60 min, followed by treatment with glutamate. Cell proliferation was measured using xCELLigence system. Treatment with all the concentrations of LP44 significantly protected the cells from the toxic effects of glutamate after 24, 48 and 72 h. Although 5-HT7 receptor expression was significantly upregulated in glutamate-treated cells, it was downregulated in LP44-pretreated cells. Furthermore, LP44 treatment significantly decreased malondialdehyde levels and increased superoxide dismutase activities and glutathione levels. Moreover, LP44 treatment significantly decreased tumor necrosis factor alpha (TNF-α) levels and inhibited caspase 3 and caspase 9 mRNA expression. In contrast, SB269970 treatment exerted an insignificant effect on oxidative stress, inflammation and apoptosis. These findings suggest that exogenous stimulation of the 5-HT7 receptors may be protective in glutamate-induced neurotoxicity and that 5-HT7 receptor agonists can be used as therapeutic agents for preventing glutamate-induced neurological disorders.
Collapse
Affiliation(s)
- Tugba Nurcan Yuksel
- Namık Kemal University, Faculty of Medicine, Department of Pharmacology, Tekirdag, Turkey
| | - Muhammed Yayla
- Kafkas University, Faculty of Medicine, Department of Pharmacology, Kars, Turkey
| | - Zekai Halici
- Ataturk University, Faculty of Medicine, Department of Pharmacology, Erzurum, Turkey.
| | - Elif Cadirci
- Ataturk University, Faculty of Medicine, Department of Pharmacology, Erzurum, Turkey
| | - Beyzagul Polat
- Ataturk University, Faculty of Pharmacy, Department of Pharmacology, Erzurum, Turkey
| | - Duygu Kose
- Ataturk University, Faculty of Medicine, Department of Pharmacology, Erzurum, Turkey
| |
Collapse
|
35
|
Mantle D, Hargreaves I. Coenzyme Q10 and Degenerative Disorders Affecting Longevity: An Overview. Antioxidants (Basel) 2019; 8:antiox8020044. [PMID: 30781472 PMCID: PMC6406788 DOI: 10.3390/antiox8020044] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/09/2019] [Accepted: 02/12/2019] [Indexed: 12/16/2022] Open
Abstract
Longevity is determined by a number of factors, including genetic, environmental and lifestyle factors. A major factor affecting longevity is the development of degenerative disorders such as cardiovascular disease, diabetes, kidney disease and liver disease, particularly where these occur as co-morbidities. In this article, we review the potential role of supplementation with coenzyme Q10 (CoQ10) for the prevention or management of these disorders. Thus, randomised controlled clinical trials have shown supplementation with CoQ10 or CoQ10 plus selenium reduces mortality by approximately 50% in patients with cardiovascular disease, or in the normal elderly population, respectively. Similarly, CoQ10 supplementation improves glycaemic control and vascular dysfunction in type II diabetes, improves renal function in patients with chronic kidney disease, and reduces liver inflammation in patients with non-alcoholic fatty liver disease. The beneficial role of supplemental CoQ10 in the above disorders is considered to result from a combination of its roles in cellular energy generation, as an antioxidant and as an anti-inflammatory agent.
Collapse
Affiliation(s)
- David Mantle
- Pharma Nord (UK) Ltd., Telford Court, Morpeth, NE61 2DB Northumberland, UK.
| | - Iain Hargreaves
- School of Pharmacy, Liverpool John Moores University, L3 5UA Liverpool, UK.
| |
Collapse
|
36
|
Casagrande D, Waib PH, Jordão Júnior AA. Mechanisms of action and effects of the administration of Coenzyme Q10 on metabolic syndrome. JOURNAL OF NUTRITION & INTERMEDIARY METABOLISM 2018. [DOI: 10.1016/j.jnim.2018.08.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
37
|
The effect of Coenzyme Q10 supplementation on serum levels of lactate, pyruvate, matrix metalloproteinase 9 and nitric oxide in women with migraine. A double blind, placebo, controlled randomized clinical trial. Eur J Integr Med 2018. [DOI: 10.1016/j.eujim.2018.06.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
38
|
Dicalcium Silicate Induced Proinflammatory Responses through TLR2-Mediated NF- κB and JNK Pathways in the Murine RAW 264.7 Macrophage Cell Line. Mediators Inflamm 2018; 2018:8167932. [PMID: 29853794 PMCID: PMC5954956 DOI: 10.1155/2018/8167932] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 03/08/2018] [Indexed: 11/21/2022] Open
Abstract
Proinflammatory responses are important aspects of the immune response to biomaterials, which may cause peri-implantitis and implant shedding. The purpose of this study was to test the cytotoxicity and proinflammatory effects of dicalcium silicate particles on RAW 264.7 macrophages and to investigate the proinflammatory response mechanism induced by C2S and tricalcium phosphate (TCP). C2S and TCP particles were characterized using scanning electron microscopy (SEM), energy spectrum analysis (EDS) and X-ray diffraction (XRD). Cytotoxicity and apoptosis assays with C2S and TCP in the murine RAW 264.7 cell line were tested using the cell counting kit-8 (CCK-8) assay and flow cytometry (FCM). The detection results showed that C2S and TCP particles had no obvious toxicity in RAW 264.7 cells and did not cause obvious apoptosis, although they both caused an oxidative stress response by producing ROS when the concentrations were at 100 μg/mL. C2S particles are likely to induce a proinflammatory response by inducing high TLR2, TNF-α mRNA, TNF-α proinflammatory cytokine, p-IκB, and p-JNK1 + JNK2 + JNK3 expression levels. When we added siRNA-TLR2-1, a significant reduction was observed. These findings support the theory that C2S particles induce proinflammatory responses through the TLR2-mediated NF-κB and JNK pathways in the murine RAW 264.7 macrophage cell line.
Collapse
|
39
|
Role of Reduced Nitric Oxide in Liver Cell Apoptosis Inhibition During Liver Damage. Arch Med Res 2018; 49:219-225. [DOI: 10.1016/j.arcmed.2018.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 09/14/2018] [Indexed: 01/05/2023]
|
40
|
Zargar S, Al-Majed ARA, Wani TA. Potentiating and synergistic effect of grapefruit juice on the antioxidant and anti-inflammatory activity of aripiprazole against hydrogen peroxide induced oxidative stress in mice. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 18:106. [PMID: 29566693 PMCID: PMC5865358 DOI: 10.1186/s12906-018-2169-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 03/15/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Dependence on antipsycotic drugs like aripriprazole (ARI) is increasing at alarming rate, hence, this study was undertaken to support the hypothesis that supplementation of Citrus paradisi (Grapefruit) juice having high concentration of polyphenols might potentiate and synergize the therapeutic effect of ARI, by increasing its bioavailability and inherent antioxidant potential. These benefits together might decrease the daily dosage of the ARI and thus alleviate the possible side effects of drug. METHODS In this study the antioxidant and anti-inflammatory potential of ARI alone and in combination with GFJ was evaluated for hydrogen peroxide (H2O2) induced oxidative stress in mice. Seventy mice (4 weeks old), were randomly divided into seven groups. Group I: Control; Group II: H2O2 treated; Group III; ARI treated; Group IV GFJ treated; Group V: GFJ and H2O2 treated; Group VI; ARI and H2O2 treated; Group VII; ARI, GFJ and H2O2 treated. Serum levels of alanine aminotransferase (ALT), blood urea nitrogen (BUN), creatinine kinase (CK), creatinine and total protein were measured. Furthermore, pro-inflammatory cytokines Interleukin (IL)-1α, IL-2, IL-10 and tumor necrosis factor-α (TNF-α) concentrations were also measured. RESULTS The mice group that was treated with ARI, GFJ or combination of the two showed significant improvement in the H2O2 altered parameters with the combination group showing more significant improvement than the ARI and GFJ alone groups indicating a synergistic and potentiating effect of the antioxidant and anti-inflammatory potential of GFJ on ARI. CONCLUSION Supplementing GFJ to ARI might increase an anti-oxidative potential of ARI due to inherent antioxidant and anti-inflammatory activity of GFJ and thus could alleviate the possible dosage dependent side effects of ARI.
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
|
Abd El-Aal SA, Abd El-Fattah MA, El-Abhar HS. CoQ10 Augments Rosuvastatin Neuroprotective Effect in a Model of Global Ischemia via Inhibition of NF-κB/JNK3/Bax and Activation of Akt/FOXO3A/Bim Cues. Front Pharmacol 2017; 8:735. [PMID: 29081748 PMCID: PMC5645536 DOI: 10.3389/fphar.2017.00735] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 09/29/2017] [Indexed: 12/12/2022] Open
Abstract
Statins were reported to lower the Coenzyme Q10 (CoQ10) content upon their inhibition of HMG-CoA reductase enzyme and both are known to possess neuroprotective potentials; therefore, the aim is to assess the possible use of CoQ10 as an adds-on therapy to rosuvastatin to improve its effect using global I/R model. Rats were allocated into sham, I/R, rosuvastatin (10 mg/kg), CoQ10 (10 mg/kg) and their combination. Drugs were administered orally for 7 days before I/R. Pretreatment with rosuvastatin and/or CoQ10 inhibited the hippocampal content of malondialdehyde, nitric oxide, and boosted glutathione and superoxide dismutase. They also opposed the upregulation of gp91phox, and p47phox subunits of NADPH oxidase. Meanwhile, both agents reduced content/expression of TNF-α, iNOS, NF-κBp65, ICAM-1, and MPO. Besides, all regimens abated cytochrome c, caspase-3 and Bax, but increased Bcl-2 in favor of cell survival. On the molecular level, they increased p-Akt and its downstream target p-FOXO3A, with the inhibition of the nuclear content of FOXO3A to downregulate the expression of Bim, a pro-apoptotic gene. Additionally, both treatments downregulate the JNK3/c-Jun signaling pathway. The effect of the combination regimen overrides that of either treatment alone. These effects were reflected on the alleviation of the hippocampal damage in CA1 region inflicted by I/R. Together, these findings accentuate the neuroprotective potentials of both treatments against global I/R by virtue of their rigorous multi-pronged actions, including suppression of hippocampal oxidative stress, inflammation, and apoptosis with the involvement of the Akt/FOXO3A/Bim and JNK3/c-Jun/Bax signaling pathways. The study also nominates CoQ10 as an adds-on therapy with statins.
Collapse
Affiliation(s)
- Sarah A Abd El-Aal
- Department of Pharmacology and Toxicology, October 6 University, Cairo, Egypt
| | - Mai A Abd El-Fattah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Hanan S El-Abhar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
43
|
Paunović MG, Matić MM, Ognjanović BI, Saičić ZS. Antioxidative and haematoprotective activity of coenzyme Q10 and vitamin E against cadmium-induced oxidative stress in Wistar rats. Toxicol Ind Health 2017; 33:746-756. [PMID: 28950793 DOI: 10.1177/0748233717725480] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Cadmium (Cd) is a major environmental pollutant, which exerts adverse effects mainly by inducing oxidative stress. Coenzyme Q10 (CoQ10) and vitamin E (VE), naturally occurring antioxidants, improve health condition by inactivating free radicals and enhancing antioxidative defence. The aim of our study was to investigate the protective role of CoQ10 and/or VE pretreatment against Cd-induced haematotoxicity. Wistar albino rats were intramuscularly injected with CoQ10 (20 mg/kg b.w.) and/or VE (20 IU/kg b.w.) or with saline (control group). After 24 h, Cd was injected intraperitoneally (0.4 mg/kg b.w.) and 1 day after, animals were sacrificed. Acute Cd intoxication caused significant changes in haematological and biochemical parameters and altered the glutathione cycle, leading to the formation of lipid peroxidation, while the concentrations and activities of antioxidants (vitamins C and E, superoxide dismutase, catalase, glutathione peroxidase and glutathione reductase) were decreased. CoQ10 and/or VE significantly maintained these values to near-normal levels, afforded additional protection by reducing lipid peroxidation and improved the levels of antioxidants in the blood. Plasma CoQ10 and VE levels negatively correlated with oxidative damage parameters while positively correlated with antioxidative defence parameters. Regarding their effects, CoQ10 and VE were in synergistic interaction. The present study suggested that CoQ10 and VE combination may be beneficial in protecting from Cd-induced haematotoxicity and may be used as a preventive against acute Cd intoxication of exposed people.
Collapse
Affiliation(s)
- Milica G Paunović
- 1 University of Kragujevac, Faculty of Science, Department of Biology and Ecology, Kragujevac, Republic of Serbia
| | - Miloš M Matić
- 1 University of Kragujevac, Faculty of Science, Department of Biology and Ecology, Kragujevac, Republic of Serbia
| | - Branka I Ognjanović
- 1 University of Kragujevac, Faculty of Science, Department of Biology and Ecology, Kragujevac, Republic of Serbia
| | - Zorica S Saičić
- 2 University of Belgrade, Institute for Biological Research "Siniša Stanković", Belgrade, Republic of Serbia
| |
Collapse
|
44
|
Khattab A, Hassanin L, Zaki N. Self-Nanoemulsifying Drug Delivery System of Coenzyme (Q10) with Improved Dissolution, Bioavailability, and Protective Efficiency on Liver Fibrosis. AAPS PharmSciTech 2017; 18:1657-1672. [PMID: 27677262 DOI: 10.1208/s12249-016-0632-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 09/10/2016] [Indexed: 01/21/2023] Open
Abstract
The aim of our investigation is to develop and characterize self-nanoemulsifying drug delivery systems (SNEDDS) of CoQ10 to improve its water solubility, dissolution rate, and bioavailability, and then evaluate its biochemical and physiological effect on liver cirrhosis in rats compared with CoQ10 powder. SNEDDS are isotropic and thermodynamically stable mixture of oil, surfactant, co-surfactant, and drug that form an oil/water nanoemulsion when added to aqueous phases with soft agitation. Upon administration, self-nanoemulsifying system becomes in contact with gastrointestinal fluid and forms o/w nanoemulsion by the aid of gastrointestinal motility. When the nanoemulsion is formed in the gastrointestinal tract, it presents the drug in a solubilized form inside small nano-sized droplets that provide a large surface area for enhancing the drug release and absorption. Solubility of CoQ10 in various oils, surfactants, and co-surfactants were studied to identify the components of SNEDDS; pseudo-ternary phase diagrams were plotted to identify the efficient self-emulsifying regions. CoQ10-loaded SNEDDS were prepared using isopropyl myristate as oil; Cremophor El, Labrasol, or Tween80 as surfactant; and Transcutol as co-surfactant. The amount of CoQ10 in each vehicle was 3%. The formulations that passed thermostability evaluation test were assessed for particle size analysis, morphological characterization, refractive index, zeta potential, viscosity, electroconductivity, drug release profile, as well as ex vivo permeability. Pharmacokinetics and hepatoprotective efficiency of the optimized SNEDDS of CoQ10 compared with CoQ10 suspension were performed. Results showed that all optimized formulae have the ability to form a good and stable nanoemulsion when diluted with water; the mean droplet size of all formulae was in the nanometric range (11.7-13.5 nm) with optimum polydispersity index values (0.2-0.21). All formulae showed negative zeta potential (-11.3 to -17.2), and maximum drug loading efficiency. One hundred percent of CoQ10 was released from most formulae within 30 min. One hundred percent of CoQ10 was permeated from all formulae through 10 h. The pharmacokinetic study in rabbits revealed a significant increase in bioavailability of CoQ10 SNEDDS to 2.1-fold compared with CoQ10 suspension after oral administration. Comparative effect of the optimized formulae on acute liver injury compared with CoQ10 powder was also studied; it was found that all the liver biochemical markers as alanine transferase (ALT), aspartate amino transferase (AST), alkaline phosphatase (ALP), total protein (TP), and albumin were significantly improved at p < 0.05. Also, histochemical and histopthological studies confirm the biochemical results. Our results suggest the potential use of SNEDDS to increase the solubility of liphophilic drug as poorly water-soluble CoQ10 and improve its oral absorption, so it can be more efficient to improve liver damage compared to CoQ10 powder. These results demonstrated that CoQ10 SNEDDS inhibited thioacetamide (TAA)-induced liver fibrosis mainly through suppression of collagen production.
Collapse
|
45
|
Chen LC, Hu LH, Yin MC. Alleviative effects from boswellic acid on acetaminophen-induced hepatic injury - Corrected and republished from: Biomedicine (Taipei). 2016 Jun; 6 (2): 9. doi: 10.7603/s40681-016-0009-1PMCID: PMC4864770. Biomedicine (Taipei) 2017; 7:13. [PMID: 28612711 PMCID: PMC5479439 DOI: 10.1051/bmdcn/2017070207] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 04/11/2016] [Indexed: 01/10/2023] Open
Abstract
Protective effects of boswellic acid (BA) against acetaminophen (APAP)-induced hepatotoxicity in Balb/ cA mice were examined. BA, at 0.05 or 0.1%, was supplied for 4 weeks. Acute liver injury was induced by APAP treatment. Results showed that BA intake increased hepatic BA bioavailability. APAP treatment decreased glutathione (GSH) level, increased reactive oxygen species (ROS) and oxidized glutathione (GSSG) production; and lowered activity and protein expression of glutathione reductase (GR) and heme oxygenase (HO)-1 in liver. BA intake at both doses alleviated subsequent APAP-induced oxidative stress by retaining GSH content, decreasing ROS and GSSG formations, reserving activity and expression of GR and HO-1 in liver, and lowering hepatic cytochrome P450 2E1 activity and expression. APAP treatment enhanced hepatic levels of interleukin-6, tumor necrosis factor-alpha and monocyte chemoattractant protein-1. BA pre-intake diminished APAP-induced release of those inflammatory cytokines and chemokines. APAP up-regulated hepatic protein expression of toll-like receptor (TLR)-3, TLR-4, MyD88, nuclear factor kappa B (NF-κB) p50, NF-κB p65 and JNK. BA pre-intake at both doses suppressed the expression of NF-κB p65 and p-JNK, and only at 0.1% down-regulated hepatic TLR-3, TLR-4 and MyD88 expression. APAP led to obvious foci of inflammatory cell infiltration in liver, determined by H&E stain. BA intake at both doses attenuated hepatic inflammatory infiltration. These findings support that boswellic acid is a potent hepato-protective agent.
Collapse
Affiliation(s)
- Lung-Che Chen
- Department of Otolaryngology, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Li-Hong Hu
- Shanghai Research Center for the Modernization of Traditional Chinese Medicine, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Mei-Chin Yin
- Department of Nutrition, China Medical University, Taichung 404, Taiwan
| |
Collapse
|
46
|
Effects of coenzyme Q 10 on the antioxidant system in SD rats exposed to lipopolysaccharide-induced toxicity. Lab Anim Res 2017; 33:24-31. [PMID: 28400836 PMCID: PMC5385279 DOI: 10.5625/lar.2017.33.1.24] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 02/22/2017] [Accepted: 03/16/2017] [Indexed: 11/21/2022] Open
Abstract
The study was performed to see the effects of coenzyme Q10 (CoQ10) on blood biochemical components and hepatic antioxidant system in rats exposed to lipopolysaccharide (LPS)-induced toxicity. A total of 24 rats were allocated to four groups: control (CON), 100 mg/kg BW of LPS (LPS), 100 mg of CoQ10/kg BW with LPS (LCQI) and 300 mg of CoQ10/kg BW with LPS (LCQII). The LPS and LCQI groups showed a significant (P<0.05) increase in the relative spleen weight compared with the CON group without affecting body and liver weights. The blood alanine aminotransferase (ALT) level in the LPS group was significantly (P<0.05) greater than that in the CON group, while supplementation with 100 or 300 mg CoQ10 to rats injected with LPS normalized the ALT level in the CON group. In antioxidant systems, the LPS group showed a significantly (P<0.05) higher mRNA and activity of superoxide dismutase (SOD) than the CON group. The supplementation with CoQ10 to the LPS-treated group normalized the level of SOD, which was comparable to the level of the CON group. Both the mRNA expression and activity of glutathione peroxidase in the LCQI and LCQII groups were higher (P<0.05) than that of the LPS group. However, administration of LPS or CoQ10 unaffected the level of catalase and total antioxidant power. The level of lipid peroxidation in the LCQII group was lower (P<0.05) than that in the LPS group. In conclusion, CoQ10 exerted its favorable effect against liver damage by modulation of antioxidant enzymes in LPS treated rats.
Collapse
|
47
|
Saleh DO, Ahmed RF, Amin MM. Modulatory role of Co-enzyme Q10 on methionine and choline deficient diet-induced non-alcoholic steatohepatitis (NASH) in albino rats. Appl Physiol Nutr Metab 2017; 42:243-249. [DOI: 10.1139/apnm-2016-0320] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The present study aimed to evaluate the hepato-protective and neuro-protective activity of Co-enzyme Q10 (CoQ10) on non-alcoholic steatohepatitis (NASH) in albino rats induced by methionine and choline-deficient (MCD) diet. Rats were fed an MCD diet for 8 weeks to induce non-alcoholic steatohepatitis. CoQ10 (10 mg/(kg·day)−1) was orally administered for 2 consecutive weeks. Twenty-four hours after the last dose of the drug, the behavioral test, namely the activity cage test, was performed and the activity counts were recorded. Serum alanine transaminase, aspartate aminotransferase, alkaline phosphatase, gamma-glutamyl transferase, total/direct bilirubin, and albumin were valued to assess liver function. Moreover, hepatic cytokines interleukin-6 as well as its modulator nuclear factor kappa-light-chain-enhancer of activated B cells were determined. In addition, brain biomarkers, viz ammonia, nitric oxide, and brain-derived neurotrophic factor (BDNF), were measured as they are reliable indices to assess brain damage. Histopathological and immunohistochemical examination of brain proliferating cell nuclear antigen in brain and liver tissues were also evaluated. Results revealed that MCD-induced NASH showed impairment in the liver functions with an increase in the liver inflammatory markers. Moreover, NASH resulted in pronounced brain dysfunction as evidenced by hyper-locomotor activity, a decrease in the BDNF level, as well as an increase in the brain nitric oxide and ammonia contents. Oral treatment of MCD-diet−fed rats with CoQ10 for 14 days showed a marked improvement in all the assigned parameters. Finally, it can be concluded that CoQ10 has a hepatoprotective and neuroprotective role in MCD-diet−induced NASH in rats.
Collapse
Affiliation(s)
- Dalia O. Saleh
- Department of Pharmacology, Medical Division, National Research Centre, 33 EL Bohouth St., Dokki, Giza 12622, Egypt
- Department of Pharmacology, Medical Division, National Research Centre, 33 EL Bohouth St., Dokki, Giza 12622, Egypt
| | - Rania F. Ahmed
- Department of Pharmacology, Medical Division, National Research Centre, 33 EL Bohouth St., Dokki, Giza 12622, Egypt
- Department of Pharmacology, Medical Division, National Research Centre, 33 EL Bohouth St., Dokki, Giza 12622, Egypt
| | - Mohamed M. Amin
- Department of Pharmacology, Medical Division, National Research Centre, 33 EL Bohouth St., Dokki, Giza 12622, Egypt
- Department of Pharmacology, Medical Division, National Research Centre, 33 EL Bohouth St., Dokki, Giza 12622, Egypt
| |
Collapse
|
48
|
Alanazi A, Algfeley SG, Al-Hosaini KA, Korashy HM, Imam F, Nagi MN. Therapeutic potential of carfilzomib, an irreversible proteasome inhibitor, against acetaminophen-induced hepatotoxicity in mice. J Biochem Mol Toxicol 2016; 31. [DOI: 10.1002/jbt.21877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 10/02/2016] [Accepted: 10/07/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Abdulrazaq Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy; King Saud University; Riyadh 11451 Kingdom of Saudi Arabia
| | - Saleh G. Algfeley
- Department of Pharmacology and Toxicology, College of Pharmacy; King Saud University; Riyadh 11451 Kingdom of Saudi Arabia
| | - Khaled A. Al-Hosaini
- Department of Pharmacology and Toxicology, College of Pharmacy; King Saud University; Riyadh 11451 Kingdom of Saudi Arabia
| | - Hesham M. Korashy
- Department of Pharmacology and Toxicology, College of Pharmacy; King Saud University; Riyadh 11451 Kingdom of Saudi Arabia
| | - Faisal Imam
- Department of Pharmacology and Toxicology, College of Pharmacy; King Saud University; Riyadh 11451 Kingdom of Saudi Arabia
| | - Mahmoud N. Nagi
- Department of Pharmacology and Toxicology, College of Pharmacy; King Saud University; Riyadh 11451 Kingdom of Saudi Arabia
| |
Collapse
|
49
|
Eftekhari A, Ahmadian E, Azarmi Y, Parvizpur A, Hamishehkar H, Eghbal MA. In vitro/vivo studies towards mechanisms of risperidone-induced oxidative stress and the protective role of coenzyme Q10 and N-acetylcysteine. Toxicol Mech Methods 2016; 26:520-528. [DOI: 10.1080/15376516.2016.1204641] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Aziz Eftekhari
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Pharmacology and Toxicology Department, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Students’ Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Ahmadian
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Pharmacology and Toxicology Department, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Azarmi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Pharmacology and Toxicology Department, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Parvizpur
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Pharmacology and Toxicology Department, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Hamishehkar
- Drug Applied Research Center, 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, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
50
|
Ashkani-Esfahani S, Bagheri F, Emami Y, Esmaeilzadeh E, Azarpira N, Hassanabadi N, Keshtkar M, Farjam M, Koohi-Hosseinabadi O, Noorafshan A. Protective Effects of Co-Enzyme Q10 on Thioacetamide-Induced Acute Liver Damage and Its Correlation With Behavioral, Biochemical, and Pathological Factors. IRANIAN RED CRESCENT MEDICAL JOURNAL 2016; 18:e29166. [PMID: 28058114 PMCID: PMC5192999 DOI: 10.5812/ircmj.29166] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 08/29/2015] [Accepted: 09/26/2015] [Indexed: 12/25/2022]
Abstract
Background Acute liver damage may be followed by biochemical, behavioral, and pathological alterations, which can result in serious complications and even death. Objectives In this experimental study we determined whether coenzyme Q10 (CoQ10), a common supplementary medicine known to have protective, antioxidative, and anti-inflammatory effects in cells, has any protective effect against thioacetamide (TAA)-induced liver damage and its related neurobehavioral alterations in rats. Materials and Methods In this experimental study forty-eight Wistar rats were divided randomly into four groups (n = 12): C1 was the control group; C2 received a single-dose of TAA (350mg/kg; intraperitoneally) without any other treatment; E1 received TAA + 5 mg/kg CoQ10 (intraperitoneally); and E2 received TAA + 10 mg/kg CoQ10. After sacrificing the rats, liver enzymes and plasma-ammonia (NH4) were measured and histopathological analyses of the livers were carried out. Elevated-plus-maze, open-field, and forced-swimming tests were also performed to investigate behavioral correlations. Results The serum levels of alanine-aminotransferase (ALT), aspartate-aminotransferase (AST), and NH4 show significant increases (P < 0.05). The groups treated with CoQ10 were shown to have significantly lower clinical grade of encephalopathy (P = 0.001), higher locomotor activity (P = 0.000), and lower levels of depression (P = 0.000). Furthermore, it was also shown that CoQ10 treatment may lead to significant decreases in scores of centrilobular necrosis, apoptosis, inflammatory cell infiltration, vacuolization, and liver necrosis (P < 0.05). Conclusions Overall, CoQ10 was determined to have positive effects on liver injury and its related behavioral and biochemical changes.
Collapse
Affiliation(s)
| | - Fereshteh Bagheri
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, IR Iran
| | - Yasaman Emami
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, IR Iran
| | - Elmira Esmaeilzadeh
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, IR Iran
| | - Negar Azarpira
- Organ Transplant Research Center, Namazi Hospital, Shiraz University of Medical Sciences, Shiraz, IR Iran
| | - Nazila Hassanabadi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, IR Iran
| | - Marzieh Keshtkar
- International Branch, Shiraz University of Medical Sciences, Kish, IR Iran
| | - Mojtaba Farjam
- Department Of Pharmacology, Fasa University of Medical Sciences, Shiraz, IR Iran
| | - Omid Koohi-Hosseinabadi
- Center of Comparative and Experimental Medicine, Shiraz University of Medical Sciences, Shiraz, IR Iran
| | - Ali Noorafshan
- Histomorphometry and Stereology Research Centre, Shiraz University of Medical Sciences, Shiraz, IR Iran
- Corresponding Author: Ali Noorafshan, Histomorphometry and Stereology Research Centre, Shiraz University of Medical Sciences, Shiraz, IR Iran. Tel: +98-9173397040, Fax: +98-7136262034, E-mail:
| |
Collapse
|