1
|
Liu H, Jiang L, Xu S, Wang C, Sun J. Quercetin prevents methylmercury-induced mitochondrial dysfunction in the cerebral cortex of mice. Drug Chem Toxicol 2024; 47:1124-1138. [PMID: 38647114 DOI: 10.1080/01480545.2024.2341888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 04/06/2024] [Indexed: 04/25/2024]
Abstract
Methylmercury (MeHg) exposure can cause nerve damage and mitochondrial dysfunction. Mitochondrial dysfunction is mainly mediated by mitochondrial biogenesis and mitochondrial dynamics disorders. Quercetin (QE) plays an important role in activating silencing information regulator 2 related enzyme 1 (SIRT1), and SIRT1 activates peroxisome-proliferator-activated receptor-γ co-activator 1α (PGC-1α), which can regulate mitochondrial biogenesis and mitochondrial dynamics. The main purpose of this study was to explore the alleviating effects of QE on MeHg-induced nerve damage and mitochondrial dysfunction. The results showed that QE could reduce the excessive production of reactive oxygen species (ROS) and the loss of membrane potential induced by MeHg. Meanwhile, QE activated SIRT1 activity and SIRT1/PGC-1α signaling pathway, improved mitochondrial biogenesis and fusion and reduced mitochondrial fission. In summary, we hypothesized that QE prevents MeHg-induced mitochondrial dysfunction by activating SIRT1/PGC-1α signaling pathway.
Collapse
Affiliation(s)
- Haihui Liu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Liujiangshan Jiang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Si Xu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Chen Wang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, PR China
| | - Jingyi Sun
- Department of Cardiology, The Second Hospital of Dalian Medical University, Dalian, PR China
| |
Collapse
|
2
|
Sedky A, Famurewa AC. Anti-ischemic drug trimetazidine blocks mercury nephrotoxicity by suppressing renal redox imbalance, inflammatory stress and caspase-dependent apoptosis in rats. Drug Chem Toxicol 2024; 47:674-681. [PMID: 37528808 DOI: 10.1080/01480545.2023.2242007] [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: 05/10/2023] [Revised: 07/12/2023] [Accepted: 07/21/2023] [Indexed: 08/03/2023]
Abstract
Trimetazidine (TMZ) is a promising emerging therapeutic piperazine derivative for renal pathologies. However, the nephroprotective mechanism of TMZ against heavy metal-induced toxicity is unknown. This study, therefore, aimed to explore whether TMZ could mitigate mercury-induced nephrotoxicity in rats. Rats were injected TMZ (3 mg/kg bw) and/or mercury chloride (HgCl2) (4 mg/kg bw) for 4 days (n = 6 rats per group). The blood analysis revealed marked increases in creatinine, urea and uric acid levels in HgCl2 group compared to the control. HgCl2 induced prominent decreases in renal superoxide dismutase (SOD), catalase (CAT), glutathione peroxide (GPx) activities compared to the control followed by marked increases in the levels of malondialdehyde (MDA), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), caspase-3 and caspase-9. Whereas the renal levels of anti-inflammatory cytokines interleukin-4 (IL-4) and interleukin-10 (IL-10) reduced considerably compared to the control. Contrarily, it was found that in the rats administered TMZ + HgCl2, levels of renal markers, MDA, TNF-α, IL-6 and caspases-3/-9 were prominently reduced compared to the HgCl2 group. The renal SOD, CAT, GPx, IL-4, and IL-10 were markedly elevated along with ameliorated histopathological lesions. On the whole, therefore, TMZ could be repurposed for blocking HgCl2 nephrotoxicity via inhibition of oxidative inflammation and apoptosis in rats.
Collapse
Affiliation(s)
- Azza Sedky
- Department of Biological Sciences, College of Science, King Faisal University, Saudi Arabia
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medical Sciences, Alex Ekwueme Federal University, Ndufu-Alike, Ikwo, Nigeria
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
3
|
Alfwuaires MA, Famurewa AC, Algefare AI, Sedky A. Naringenin blocks hepatic cadmium accumulation and suppresses cadmium-induced hepatotoxicity via amelioration of oxidative inflammatory signaling and apoptosis in rats. Drug Chem Toxicol 2024; 47:436-444. [PMID: 37073537 DOI: 10.1080/01480545.2023.2196377] [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: 12/19/2022] [Accepted: 03/22/2023] [Indexed: 04/20/2023]
Abstract
Liver is one of the targets of cadmium (Cd) bioaccumulation for hepatic damage and pathologies via oxidative inflammation and apoptosis. The current study explored whether the citrus flavonoid naringenin (NAR) could prevent hepatic accumulation of Cd and Cd hepatotoxicity in a rat model. Rats in group 1 received normal saline; group 2 received NAR (50 mg/kg body weight); group 3 received CdCl2 (5 mg/kg body weight); group 4 received NAR + CdCl2, for four consecutive weeks. Assays related to markers of oxidative stress, inflammation, and apoptosis were carried out using liver homogenate. Blood and liver sample analyses revealed significant elevation of blood and hepatic Cd levels coupled with prominent increases in alkaline phosphatase (ALP), alanine aminotransferase (ALT), and aspartate aminotransferase (AST) activities, whereas the albumin and total protein levels were decreased considerably. Hepatic superoxide dismutase (SOD), catalase (CAT), glutathione peroxide (GPx) activities diminished significantly compared to control followed by marked increases in malondialdehyde (MDA) levels, and dysregulation in caspase and cytokine (TNF-α, IL-6, IL-4, IL-10) levels. However, it was found that in the rats administered NAR + Cd, the levels of Cd, hepatic enzymes, MDA, TNF-α, IL-6, and caspases-3/-9 were prominently reduced compared to the Cd group. The hepatic SOD, CAT, GPx, IL-4, IL-10, albumin, and total protein were markedly elevated along with alleviated hepatic histopathological abrasions. Taken together therefore, NAR is a potential flavonoid for blocking hepatic Cd bioaccumulation and consequent inhibition of Cd-induced oxidative inflammation and apoptotic effects on the liver of rats.
Collapse
Affiliation(s)
- Manal A Alfwuaires
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical, Sciences, College of Medical Sciences, Alex-Ekwueme Federal University, Ndufu-Alike, Ikwo, Nigeria
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Abdulmohsen I Algefare
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Azza Sedky
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| |
Collapse
|
4
|
Kahramanoğullari M, Erişir M, Yaman M, Parlak Ak T. Effects of naringenin on oxidative damage and apoptosis in liver and kidney in rats subjected to chronic mercury chloride. ENVIRONMENTAL TOXICOLOGY 2024; 39:2937-2947. [PMID: 38308452 DOI: 10.1002/tox.24164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/03/2024] [Accepted: 01/18/2024] [Indexed: 02/04/2024]
Abstract
Mercury chloride is a type of heavy metal that causes the formation of free radicals, causing hepatotoxicity, nephrotoxicity and apoptosis. In this study, the effects of naringenin on oxidative stress and apoptosis in the liver and kidney of rats exposed to mercury chloride were investigated. In the study, 41 2-month-old male Wistar-Albino rats were divided into five groups. Accordingly, group 1 was set as control group, group 2 as naringenin-100, group 3 as mercury chloride, group 4 as mercury chloride + naringenin-50, and group 5 as mercury chloride + naringenin-100. For the interventions, 1 mL/kg saline was administered to the control, 0.4 mg/kg/day mercury (II) chloride to the mercury chloride groups by i.p., and 50 and 100 mg/kg/day naringenin prepared in corn oil to the naringenin groups by gavage. All the interventions lasted for 20 days. Mercury chloride administration was initiated 1 h following the administration of naringenin. When mercury chloride and the control group were compared, a significant increase in plasma urea, liver and kidney malondialdehyde (MDA) levels, in kidney superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), glutathione-S-transferase (GST) activities (p < .001), and a significant decrease in liver and kidney glutathione (GSH) levels (p < .001), in liver catalase (CAT) activity (p < .01) were observed. In addition, histopathological changes and a significant increase in caspase-3 levels were detected (p < .05). When mercury chloride and treatment groups were compared, the administration of naringenin caused a decrease aspartate transaminase (AST), alanine transaminase (ALT), lactate dehydrogenase (LDH) (p < .01), urea, creatinine levels (p < .001) in plasma, MDA levels in liver and kidney, SOD, GSH-Px, GST activities in kidney (p < .001), and increased GSH levels in liver and kidney. The addition of naringenin-100 increased GSH levels above the control (p < .001). The administration of naringenin was also decreased histopathological changes and caspase-3 levels (p < .05). Accordingly, it was determined that naringenin is protective and therapeutic against mercury chloride-induced oxidative damage and apoptosis in the liver and kidney, and 100 mg/kg naringenin is more effective in preventing histopathological changes and apoptosis.
Collapse
Affiliation(s)
- Merve Kahramanoğullari
- Department of Biochemistry, Faculty of Veterinary Medicine, Fırat University, Elazığ, Turkey
| | - Mine Erişir
- Department of Biochemistry, Faculty of Veterinary Medicine, Fırat University, Elazığ, Turkey
| | - Mine Yaman
- Department of Histology-Embryology, Faculty of Veterinary Medicine, Fırat University, Elazığ, Turkey
| | - Tuba Parlak Ak
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Munzur University, Tunceli, Turkey
| |
Collapse
|
5
|
Zhu XQ, Gao D. Naringenin alleviates cognitive dysfunction in rats with cerebral ischemia/reperfusion injury through up-regulating hippocampal BDNF-TrkB signaling: involving suppression in neuroinflammation and oxidative stress. Neuroreport 2024; 35:216-224. [PMID: 38141009 PMCID: PMC10852040 DOI: 10.1097/wnr.0000000000001989] [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: 10/29/2023] [Accepted: 12/03/2023] [Indexed: 12/24/2023]
Abstract
Cognitive dysfunction is one of the common complications of cerebral ischemia-reperfusion (CI/R) injury after ischemic stroke. Neuroinflammation and oxidative stress are the core pathological mechanism of CI/R injury. The activation of brain derived neurotrophic factor (BDNF)-tyrosine receptor kinase B (TrkB) signaling antagonize cognitive dysfunction in a series of neuropathy. Naringenin (NAR) improves cognitive function in many diseases, but the role of NAR in CI/R injury-induced cognitive dysfunction remains unexplored. The study aimed to explore the potential protective effects of NAR in CI/R injury-induced cognitive dysfunction and underlying mechanism. The rats were exposed to transient middle cerebral artery occlusion (MCAO) and then treated with distilled water or NAR (50 or 100 mg/kg/day, p.o.) for 30 days. The Y-maze test, Novel object recognition test and Morris water maze test were performed to assess cognitive function. The levels of oxidative stress and inflammatory cytokines were measured by ELISA. The expressions of BDNF/TrkB signaling were detected by Western blot. NAR prevented cognitive impairment in MCAO-induced CI/R injury rats. Moreover, NAR inhibited oxidative stress (reduced levels of malondialdehyde and 4-hydroxynonenal, increased activities of superoxide dismutase and Glutathione peroxidase) and inflammatory cytokines (reduced levels of tumor necrosis factor-α, Interleukin-1β and Interleukin-6), up-regulated the expressions of BDNF and p-TrkB in hippocampus of MCAO-induced CI/R rats. NAR ameliorated cognitive dysfunction of CI/R rats via inhibiting oxidative stress, reducing inflammatory response, and up-regulating BDNF/TrkB signaling pathways in the hippocampus.
Collapse
Affiliation(s)
- Xiao-Qin Zhu
- Health School of Nuclear Industry, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China
| | - Dong Gao
- The Affiliated Nanhua Hospital, Medical administration division, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
6
|
Xu S, Liu H, Wang C, Deng Y, Xu B, Yang T, Liu W. Study of ATF4/CHOP axis-mediated mitochondrial unfolded protein response in neuronal apoptosis induced by methylmercury. Food Chem Toxicol 2023; 182:114190. [PMID: 37967789 DOI: 10.1016/j.fct.2023.114190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/18/2023] [Accepted: 11/08/2023] [Indexed: 11/17/2023]
Abstract
Methylmercury (MeHg) is a widely distributed environmental pollutant that can easily cross the blood-brain barrier and accumulate in the brain, thereby damaging the central nervous system. Studies have shown that MeHg-induced mitochondrial damage and apoptosis play a crucial role in its neurotoxic effects. Mitochondrial unfolded protein response (UPRmt) is indispensable to maintain mitochondrial protein homeostasis and ensure mitochondrial function, and the ATF4/CHOP axis is one of the signaling pathways to activate UPRmt. In this study, the role of the ATF4/CHOP axis-mediated UPRmt in the neurotoxicity of MeHg has been investigated by C57BL/6 mice and the HT22 cell line. We discovered that mice exposed to MeHg had abnormal neurobehavioral patterns. The pathological section showed a significant decrease in the number of neurons. MeHg also resulted in a reduction in mtDNA copy number and mitochondrial membrane potential (MMP). Additionally, the ATF4/CHOP axis and UPRmt were found to be significantly activated. Subsequently, we used siRNA to knock down ATF4 or CHOP and observed that the expression of UPRmt-related proteins and the apoptosis rate were significantly reduced. Our research showed that exposure to MeHg can over-activate the UPRmt through the ATF4/CHOP axis, leading to mitochondrial damage and ultimately inducing neuronal apoptosis.
Collapse
Affiliation(s)
- Si Xu
- Department of Environmental Health, School of Public Health, China Medical University, China
| | - Haihui Liu
- Department of Environmental Health, School of Public Health, China Medical University, China
| | - Chen Wang
- Department of Environmental Health, School of Public Health, China Medical University, China
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, China
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, China
| | - Tianyao Yang
- Department of Environmental Health, School of Public Health, China Medical University, China.
| | - Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, China.
| |
Collapse
|
7
|
Zuo K, Xu Q, Wang Y, Sui Y, Niu Y, Liu Z, Liu M, Liu X, Liu D, Sun W, Wang Z, Liu X, Liu J. L-Ascorbic Acid 2-Phosphate Attenuates Methylmercury-Induced Apoptosis by Inhibiting Reactive Oxygen Species Accumulation and DNA Damage in Human SH-SY5Y Cells. TOXICS 2023; 11:144. [PMID: 36851019 PMCID: PMC9967424 DOI: 10.3390/toxics11020144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/28/2023] [Accepted: 01/31/2023] [Indexed: 06/18/2023]
Abstract
Methylmercury (MeHg) is a toxin that causes severe neuronal oxidative damage. As vitamin C is an antioxidant well-known to protect neurons from oxidative damage, our goal was to elucidate its protective mechanism against MeHg-induced oxidative stress in human neuroblastomas (SHSY5Y). We treated cells with MeHg, L-ascorbic acid 2-phosphate (AA2P), or both, and used MTT, flow cytometry, and Western blot analyses to assess cell damage. We found that MeHg significantly decreased the survival rate of SH-SY5Y cells in a time- and dose-dependent manner, increased apoptosis, downregulated PAR and PARP1 expression, and upregulated AIF, Cyto C, and cleaved Caspase-3 expression. A time course study showed that MeHg increased reactive oxygen species (ROS) accumulation; enhanced apoptosis; increased DNA damage; upregulated expression ofγH2A.X, KU70, 67 and 57 kDa AIF, CytoC, and cleaved Caspase-3; and downregulated expression of 116 kDa PARP1, PAR, BRAC1, and Rad51. Supplementation with AA2P significantly increased cell viability and decreased intrinsic ROS accumulation. It also reduced ROS accumulation in cells treated with MeHg and decreased MeHg-induced apoptosis. Furthermore, AA2P conversely regulated gene expression compared to MeHg. Collectively, we demonstrate that AA2P attenuates MeHg-induced apoptosis by alleviating ROS-mediated DNA damage and is a potential treatment for MeHg neurotoxicity.
Collapse
Affiliation(s)
- Kuiyang Zuo
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| | - Qi Xu
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| | - Yujie Wang
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| | - Yutong Sui
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| | - Ye Niu
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| | - Zinan Liu
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| | - Mingsheng Liu
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| | - Xinpeng Liu
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| | - Dan Liu
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| | - Wei Sun
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| | - Ziyu Wang
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| | - Xiaomei Liu
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| | - Jinyu Liu
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun 130021, China
| |
Collapse
|
8
|
Zhu N, Liu R, Xu MH, Li Y. Neuroprotective Actions of Different Exogenous Nucleotides in H 2O 2-Induced Cell Death in PC-12 Cells. Molecules 2023; 28:molecules28031226. [PMID: 36770893 PMCID: PMC9920452 DOI: 10.3390/molecules28031226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/05/2023] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
Exogenous nucleotides (NTs) are considered conditionally essential nutrients, and the brain cannot synthesize NTs de novo. Therefore, the external supplementation of exogenous NTs is of great significance for maintaining normal neuronal metabolism and function under certain conditions, such as brain aging. This study, therefore, sets out to assess the neuroprotective effect of four kinds of single exogenous NTs and a mixture of the NTs, and to elucidate the potential mechanism. A rat pheochromocytoma cell line PC-12 was treated with different concentrations of exogenous NTs after 4 h of exposure to 200 µM H2O2. We found that the exogenous NTs exerted significant neuroprotection through decreasing neuron apoptosis and DNA damage, ameliorating inflammation and mitochondrial dysfunction, promoting cell viability, and augmenting antioxidant activity, and that they tended to up-regulate the NAD+/SIRTI/PGC-1α pathway involved in mitochondrial biogenesis. Among the different NTs, the neuroprotective effect of AMP seemed to be more prominent, followed by the NT mixture, NMN, and CMP. AMP also exhibited the strongest antioxidant activity in H2O2-treated PC-12 cells. UMP was excellent at inhibiting neuronal inflammation and improving mitochondrial function, while GMP offered major advantages in stabilizing mitochondrial membrane potential. The mixture of NTs had a slightly better performance than NMN, especially in up-modulating the NAD+/SIRTI/PGC-1α pathway, which regulates mitochondrial biogenesis. These results suggest that antioxidant activity, anti-inflammatory activity, and protection of mitochondrial function are possible mechanisms of the neuroprotective actions of exogenous NTs, and that the optimization of the mixture ratio and the concentration of NTs may achieve a better outcome.
Collapse
Affiliation(s)
- Na Zhu
- Department of Nutrition and Food Hygiene, College of Public Health, Inner Mongolia Medical University, Hohhot 010059, China
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
| | - Riu Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
| | - Mei-Hong Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Yong Li
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
- Correspondence: ; Tel.: +86-10-8280-1177
| |
Collapse
|
9
|
Galluzzi S, Zanardini R, Ferrari C, Gipponi S, Passeggia I, Rampini M, Sgrò G, Genovese S, Fiorito S, Palumbo L, Pievani M, Frisoni GB, Epifano F. Cognitive and biological effects of citrus phytochemicals in subjective cognitive decline: a 36-week, randomized, placebo-controlled trial. Nutr J 2022; 21:64. [PMID: 36253765 PMCID: PMC9575277 DOI: 10.1186/s12937-022-00817-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 09/28/2022] [Indexed: 11/10/2022] Open
Abstract
Background Auraptene (AUR) and naringenin (NAR) are citrus-derived phytochemicals that influence several biological mechanisms associated with cognitive decline, including neuronal damage, oxidative stress and inflammation. Clinical evidence of the efficacy of a nutraceutical with the potential to enhance cognitive function in cohorts at risk of cognitive decline would be of great value from a preventive perspective. The primary aim of this study is to determine the cognitive effects of a 36-week treatment with citrus peel extract standardized in levels of AUR and NAR in older adults experiencing subjective cognitive decline (SCD). The secondary aim is to determine the effects of these phytochemicals on blood-based biomarkers indicative of neuronal damage, oxidative stress, and inflammation. Methods Eighty older persons with SCD will be recruited and randomly assigned to receive the active treatment (400 mg of citrus peel extract containing 0.1 mg of AUR and 3 mg of NAR) or the placebo at a 1:1 ratio for 36 weeks. The primary endpoint is a change in the Repeatable Battery for the Assessment of Neuropsychological Status score from baseline to weeks 18 and 36. Other cognitive outcomes will include changes in verbal and nonverbal memory, attention, executive and visuospatial functions. Blood samples will be collected from a consecutive subsample of 60 participants. The secondary endpoint is a change in interleukin-8 levels over the 36-week period. Other biological outcomes include changes in markers of neuronal damage, oxidative stress, and pro- and anti-inflammatory cytokines. Conclusion This study will evaluate whether an intervention with citrus peel extract standardized in levels of AUR and NAR has cognitive and biological effects in older adults with SCD, facilitating the establishment of nutrition intervention in people at risk of cognitive decline. Trial registration The trial is registered with the United States National Library of Medicine at the National Institutes of Health Registry of Clinical Trials under the code NCT04744922 on February 9th, 2021 (https://www.clinicaltrials.gov/ct2/show/NCT04744922).
Collapse
Affiliation(s)
- Samantha Galluzzi
- Laboratory Alzheimer's Neuroimaging and Epidemiology, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy.
| | - Roberta Zanardini
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy
| | - Clarissa Ferrari
- Service of Statistics, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy
| | - Sara Gipponi
- Laboratory Alzheimer's Neuroimaging and Epidemiology, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy
| | - Ilaria Passeggia
- Laboratory Alzheimer's Neuroimaging and Epidemiology, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy
| | - Michela Rampini
- Laboratory Alzheimer's Neuroimaging and Epidemiology, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy
| | - Giovanni Sgrò
- Clinical Trial Service, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy
| | - Salvatore Genovese
- Laboratory of Phytochemistry and Chemistry of Natural Products, Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Serena Fiorito
- Laboratory of Phytochemistry and Chemistry of Natural Products, Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Lucia Palumbo
- Laboratory of Phytochemistry and Chemistry of Natural Products, Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Michela Pievani
- Laboratory Alzheimer's Neuroimaging and Epidemiology, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy
| | | | - Francesco Epifano
- Laboratory of Phytochemistry and Chemistry of Natural Products, Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
10
|
Aoiadni N, Jdidi H, Feki AE, Fetoui H, Koubaa FG. Mitochondrial bioenergetics and redox dysfunction in nephrotoxicity induced by pyrethroid permethrin are ameliorated by flavonoid-rich fraction. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:63973-63987. [PMID: 35469380 DOI: 10.1007/s11356-022-20350-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 04/15/2022] [Indexed: 06/14/2023]
Abstract
The present study was designed to evaluate in vitro and in vivo the potential anti-inflammatory and nephroprotective potential of ethyl acetate fraction extracted from Fumaria officinalis (EAF) against permethrin (PER). Male wistar rats were treated daily by gavage during 7 days as follows: group C: negative control rats received 2 mL/kg bw of corn oil, group EAF: positive control rats received EAF at a dose of 200 mg/kg bw dissolved in water, group PER: rats received PER at a dose of 34.05 mg/kg bw and group (PER + EAF): rats received PER (34.05 mg/kg bw) and EAF (200 mg/kg bw). In vitro study showed the ability of EAF to inhibit protein denaturation and heat-induced hemolysis confirming its anti-inflammatory activity. In vivo, PER treatment decreased calcium (Ca) and phosphorus (P) levels and increased lactate dehydrogenase (LDH) activity in plasma. It induced oxidative stress objectified by an increase in the lipid peroxidation and protein oxidation and a perturbation of antioxidant system in kidney and mitochondria. The activities of NADH-ubiquinone reductase, ubiquinol-cytochrome C reductase and cytochrome C oxidase activities were reduced. These alterations were confirmed by histopathological studies. Co-treatment with EAF improved the antioxidant status and mitochondrial bioenergetics. The nephroprotective effects of EAF could be attributed to its modulation of detoxification enzymes and/or free radical scavenging actions.
Collapse
Affiliation(s)
- Nissaf Aoiadni
- Laboratory of Animal Eco-Physiology, Faculty of Sciences of Sfax, Tunisia, Street of Soukra Km 3.5, BP 1171, 9 3000, Sfax, CP, Tunisia.
| | - Hajer Jdidi
- Laboratory of Animal Eco-Physiology, Faculty of Sciences of Sfax, Tunisia, Street of Soukra Km 3.5, BP 1171, 9 3000, Sfax, CP, Tunisia
| | - Abdelfattah El Feki
- Laboratory of Animal Eco-Physiology, Faculty of Sciences of Sfax, Tunisia, Street of Soukra Km 3.5, BP 1171, 9 3000, Sfax, CP, Tunisia
| | - Hamadi Fetoui
- Laboratory of Toxicology and Environmental Health.LR17ES06, Sciences Faculty of Sfax, University of Sfax, 14 BP1171, 3000, Sfax, Tunisia
| | - Fatma Ghorbel Koubaa
- Laboratory of Animal Eco-Physiology, Faculty of Sciences of Sfax, Tunisia, Street of Soukra Km 3.5, BP 1171, 9 3000, Sfax, CP, Tunisia
| |
Collapse
|
11
|
Zhang J, Zhang Y, Liu Y, Niu X. Naringenin Attenuates Cognitive Impairment in a Rat Model of Vascular Dementia by Inhibiting Hippocampal Oxidative Stress and Inflammatory Response and Promoting N-Methyl-D-Aspartate Receptor Signaling Pathway. Neurochem Res 2022; 47:3402-3413. [PMID: 36028734 DOI: 10.1007/s11064-022-03696-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 07/11/2022] [Accepted: 07/14/2022] [Indexed: 10/15/2022]
Abstract
Vascular dementia (VaD) is the second most common form of dementia globally, yet there are no efficient treatments. Naringenin, a natural flavonoid, exerts antioxidative, anti-inflammatory, and neuroprotective properties; however, its potential effect on VaD remain unclear. Herein, the purpose of present study was to elucidate whether naringenin attenuates cognitive dysfunction in VaD via inhibiting hippocampal oxidative stress and inflammatory response, and promoting N-methyl-D-aspartate receptors (NMDARs) signaling pathway. A rat model of VaD was established by permanent bilateral common carotid artery occlusion [2-vessel occlusion (2VO)]. Behavioral performance analyses results revealed that administration of naringenin improves cognitive impairment in rats with VaD according to the new object recognition test and the Morris water maze test. In addition, naringenin attenuated hippocampal oxidative stress by reducing reactive oxygen species generation, decreasing malondialdehyde content and recombinant reactive oxygen species modulator 1 (Romo-1) expression, and increasing superoxide dismutase and glutathione peroxidase activities in the hippocampus of VaD rats. Moreover, naringenin decreased the proinflammatory cytokines (IL-1β, IL-6, and TNF-α) levels and increased the anti-inflammatory cytokines (IL-10 and IL-4) levels in the hippocampus of 2VO surgery-treated rats, attenuating hippocampal inflammatory response during VaD. Furthermore, naringenin promoted synaptophysin (SYP), postsynaptic density protein 95 (PSD95), N-methyl-Daspartic acid receptor 1 (NR1) and N-methyl-D-aspartate receptor subunit 2B (NR2B) expressions levels in hippocampus of VaD rats. Collectively, these findings indicated that naringenin mitigates cognitive impairment in VaD rats partly via inhibiting hippocampal oxidative stress and inflammatory response and restoring NMDARs signaling pathway.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Neurology, The First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Yingze District, Taiyuan, 030000, Shanxi, People's Republic of China
| | - Yu Zhang
- Department of Neurology, Shanxi Hospital of Integrated Traditional and Western Medicine, Taiyuan, 030000, Shanxi, People's Republic of China
| | - Yan Liu
- Department of Neurology, The First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Yingze District, Taiyuan, 030000, Shanxi, People's Republic of China
| | - Xiaoyuan Niu
- Department of Neurology, The First Hospital of Shanxi Medical University, No. 85 Jiefang South Road, Yingze District, Taiyuan, 030000, Shanxi, People's Republic of China.
| |
Collapse
|
12
|
Fakhri S, Sabouri S, Kiani A, Farzaei MH, Rashidi K, Mohammadi-Farani A, Mohammadi-Noori E, Abbaszadeh F. Intrathecal administration of naringenin improves motor dysfunction and neuropathic pain following compression spinal cord injury in rats: relevance to its antioxidant and anti-inflammatory activities. Korean J Pain 2022; 35:291-302. [PMID: 35768984 PMCID: PMC9251389 DOI: 10.3344/kjp.2022.35.3.291] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/08/2022] [Accepted: 05/13/2022] [Indexed: 12/02/2022] Open
Abstract
Background Spinal cord injury (SCI) is one of the most debilitating disorders throughout the world, causing persistent sensory-motor dysfunction, with no effective treatment. Oxidative stress and inflammatory responses play key roles in the secondary phase of SCI. Naringenin (NAR) is a natural flavonoid with known anti-inflammatory and antioxidative properties. This study aims at evaluating the effects of intrathecal NAR administration on sensory-motor disability after SCI. Methods Animals underwent a severe compression injury using an aneurysm clip. About 30 minutes after surgery, NAR was injected intrathecally at the doses of 5, 10, and 15 mM in 20 µL volumes. For the assessment of neuropathic pain and locomotor function, acetone drop, hot plate, inclined plane, and Basso, Beattie, Bresnahan tests were carried out weekly till day 28 post-SCI. Effects of NAR on matrix metalloproteinase (MMP)-2 and MMP-9 activity was appraised by gelatin zymography. Also, histopathological analyses and serum levels of glutathione (GSH), catalase and nitrite were measured in different groups. Results NAR reduced neuropathic pain, improved locomotor function, and also attenuated SCI-induced weight loss weekly till day 28 post-SCI. Zymography analysis showed that NAR suppressed MMP-9 activity, whereas it increased that of MMP-2, indicating its anti-neuroinflammatory effects. Also, intrathecal NAR modified oxidative stress related markers GSH, catalase, and nitrite levels. Besides, the neuroprotective effect of NAR was corroborated through increased survival of sensory and motor neurons after SCI. Conclusions These results suggest intrathecal NAR as a promising candidate for medical therapeutics for SCI-induced sensory and motor dysfunction.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shahryar Sabouri
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amir Kiani
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Khodabakhsh Rashidi
- Research Center of Oils and Fats, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ahmad Mohammadi-Farani
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ehsan Mohammadi-Noori
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Abbaszadeh
- Department of Neuroscience, Faculty of Advanced Technologies in Medical Sciences, Iran University of Medical Sciences, Tehran, Iran.,Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Liu H, Zhang C, Wen F, Feng L, Wang H, Wang W, Li P. Effects of Low-dose Mercury Exposure in Newborns on mRNA Expression Profiles. BULLETIN OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2021; 107:975-981. [PMID: 33944966 DOI: 10.1007/s00128-021-03249-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/21/2021] [Indexed: 06/12/2023]
Abstract
This study was designed to investigate the molecular mechanism of mercury (Hg) toxicity in the newborns by mRNA sequencing (mRNA-seq). A questionnaire survey, routine blood parameters of pregnant women, and umbilical cord blood (UCB) of newborns were collected. The median (25th percentile, 75th percentile) of total Hg (THg) concentrations in UCB of newborns was 3.63 (2.50, 6.19) µg/L. A total of 504 differentially expressed genes of mRNA were revealed between the case and control group, including 456 upregulated and 48 downregulated genes. The Gene Ontology (GO) analysis showed that differentially expressed genes were primarily involved in mitophagy, hemoglobin complex, and oxygen carrier activity. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis demonstrated that the most differentially expressed genes were annotated in Huntington's disease, Parkinson's disease, and Alzheimer's disease. The qRT-PCR was used to validate the results of mRNA-seq. Low-dose Hg exposure could increase blood NE# and WBC in the pregnant women. This study provides scientific evidences on mechanism of Hg toxicity in newborns.
Collapse
Affiliation(s)
- Haohao Liu
- School of Public Health/Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, 550025, Guiyang, China
| | - Chanchan Zhang
- School of Public Health/Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, 550025, Guiyang, China
| | - Fuli Wen
- School of Public Health/Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, 550025, Guiyang, China
| | - Lin Feng
- School of Public Health/Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, 550025, Guiyang, China
| | - Huiqun Wang
- School of Public Health/Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, 550025, Guiyang, China
| | - Wenjuan Wang
- School of Public Health/Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, 550025, Guiyang, China
| | - Ping Li
- School of Public Health/Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, 550025, Guiyang, China.
- State Key Laboratory of Environmental Geochemistry, Institute of Geochemistry, Chinese Academy of Sciences, 550081, Guiyang, China.
| |
Collapse
|
14
|
Nanonaringenin and Vitamin E Ameliorate Some Behavioral, Biochemical, and Brain Tissue Alterations Induced by Nicotine in Rats. J Toxicol 2021; 2021:4411316. [PMID: 34608387 PMCID: PMC8487377 DOI: 10.1155/2021/4411316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 09/11/2021] [Indexed: 02/06/2023] Open
Abstract
Nicotine is the major alkaloid present in cigarettes that induces various biochemical and behavioral changes. Nanonaringenin (NNG) and vitamin E are antioxidants that are reported to mitigate serious impairments caused by some toxins and oxidants. Thus, we aimed to investigate the efficacy of NNG, vitamin E, and their combinations to ameliorate behavioral, biochemical, and histological alterations induced by nicotine in rats. Adult male albino rats were randomly grouped into six equal groups (10 rats/group): control, N (nicotine 1 mg/kg b.w./day S/C from 15th to 45th day, 5 days a week), NNG (25 mg/kg b.w./day orally for 45 days), N + NNG, N + E (nicotine + vitamin E 200 mg/kg b.w./day orally), and N + NNG + E (nicotine + NNG + vitamin E at the aforementioned doses). Behavioral tests were conducted on day 15 and 30 postnicotine injection, while memory tests, brain neurotransmitters, antioxidants, and histopathological examination were examined at day 30 only. As a result, nicotine impaired rats' activity (hypoactivity and hyperactivity) and memory, induced anxiolytic and anxiogenic effects on rats, and altered neurotransmitters (acetylcholinesterase, serotonin, and dopamine), and redox markers (MDA, H2O2, GSH, and catalase) levels in brain homogenates. Thickening and congestion of the meninges and degeneration of the cerebral neurons and glia cells were observed. Cosupplementation with NNG, vitamin E, and their combination with nicotine was beneficial in the alleviation of activity impairments and improved short memory and cognition defects and exploratory behaviors. Our results indicate the antioxidant potential of NNG and vitamin E by modulating redox markers and neurotransmitters in the brain. Thus, data suggest that the prophylactic use of NNG, vitamin E, and/or their combination for (45 days) may have a successful amelioration of the disrupted behavior and cognition and biochemical and histopathological alterations induced by nicotine.
Collapse
|
15
|
Wei Y, Ni L, Pan J, Li X, Xu B, Deng Y, Yang T, Liu W. The Roles of Oxidative Stress in Regulating Autophagy in Methylmercury-induced Neurotoxicity. Neuroscience 2021; 469:175-190. [PMID: 34174372 DOI: 10.1016/j.neuroscience.2021.06.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 12/24/2022]
Abstract
Methylmercury (MeHg) is a potential neurotoxin that is highly toxic to the human central nervous system. Although MeHg neurotoxicity has been widely studied, the mechanism of MeHg neurotoxicity has not yet been fully elucidated. Some research evidence suggests that oxidative stress and autophagy are important molecular mechanisms of MeHg-induced neurotoxicity. Researchers have widely accepted that oxidative stress regulates the autophagy pathway. The current study reviews the activation of Nuclear factor-erythroid-2-related factor (Nrf2)-related oxidative stress pathways and autophagy signaling pathways in the case of MeHg neurotoxicity. In addition, autophagy mainly plays a role in the neurotoxicity of MeHg through mTOR-dependent and mTOR-independent autophagy signaling pathways. Finally, the regulation of autophagy by reactive oxygen species (ROS) and Nrf2 in MeHg neurotoxicity was explored in this review, providing a new concept for the study of the neurotoxicity mechanism of MeHg.
Collapse
Affiliation(s)
- Yanfeng Wei
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China
| | - Linlin Ni
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China
| | - Jingjing Pan
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China
| | - Xiaoyang Li
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China
| | - Tianyao Yang
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China
| | - Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, China.
| |
Collapse
|
16
|
Novo JP, Martins B, Raposo RS, Pereira FC, Oriá RB, Malva JO, Fontes-Ribeiro C. Cellular and Molecular Mechanisms Mediating Methylmercury Neurotoxicity and Neuroinflammation. Int J Mol Sci 2021; 22:ijms22063101. [PMID: 33803585 PMCID: PMC8003103 DOI: 10.3390/ijms22063101] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/06/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023] Open
Abstract
Methylmercury (MeHg) toxicity is a major environmental concern. In the aquatic reservoir, MeHg bioaccumulates along the food chain until it is consumed by riverine populations. There has been much interest in the neurotoxicity of MeHg due to recent environmental disasters. Studies have also addressed the implications of long-term MeHg exposure for humans. The central nervous system is particularly susceptible to the deleterious effects of MeHg, as evidenced by clinical symptoms and histopathological changes in poisoned humans. In vitro and in vivo studies have been crucial in deciphering the molecular mechanisms underlying MeHg-induced neurotoxicity. A collection of cellular and molecular alterations including cytokine release, oxidative stress, mitochondrial dysfunction, Ca2+ and glutamate dyshomeostasis, and cell death mechanisms are important consequences of brain cells exposure to MeHg. The purpose of this review is to organize an overview of the mercury cycle and MeHg poisoning events and to summarize data from cellular, animal, and human studies focusing on MeHg effects in neurons and glial cells. This review proposes an up-to-date compendium that will serve as a starting point for further studies and a consultation reference of published studies.
Collapse
Affiliation(s)
- João P. Novo
- Institute for Clinical and Biomedical Research (iCBR), Center for Innovative Biomedicine and Biotechnology (CIBB), and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.P.N.); (B.M.); (R.S.R.); (F.C.P.)
| | - Beatriz Martins
- Institute for Clinical and Biomedical Research (iCBR), Center for Innovative Biomedicine and Biotechnology (CIBB), and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.P.N.); (B.M.); (R.S.R.); (F.C.P.)
| | - Ramon S. Raposo
- Institute for Clinical and Biomedical Research (iCBR), Center for Innovative Biomedicine and Biotechnology (CIBB), and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.P.N.); (B.M.); (R.S.R.); (F.C.P.)
- Experimental Biology Core, University of Fortaleza, Health Sciences, Fortaleza 60110-001, Brazil
| | - Frederico C. Pereira
- Institute for Clinical and Biomedical Research (iCBR), Center for Innovative Biomedicine and Biotechnology (CIBB), and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.P.N.); (B.M.); (R.S.R.); (F.C.P.)
| | - Reinaldo B. Oriá
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and Institute of Biomedicine, School of Medicine, Federal University of Ceará, Fortaleza 60430-270, Brazil;
| | - João O. Malva
- Institute for Clinical and Biomedical Research (iCBR), Center for Innovative Biomedicine and Biotechnology (CIBB), and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.P.N.); (B.M.); (R.S.R.); (F.C.P.)
- Correspondence: (J.O.M.); (C.F.-R.)
| | - Carlos Fontes-Ribeiro
- Institute for Clinical and Biomedical Research (iCBR), Center for Innovative Biomedicine and Biotechnology (CIBB), and Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.P.N.); (B.M.); (R.S.R.); (F.C.P.)
- Correspondence: (J.O.M.); (C.F.-R.)
| |
Collapse
|
17
|
Naraki K, Rezaee R, Karimi G. A review on the protective effects of naringenin against natural and chemical toxic agents. Phytother Res 2021; 35:4075-4091. [PMID: 33724584 DOI: 10.1002/ptr.7071] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/13/2021] [Accepted: 02/23/2021] [Indexed: 12/14/2022]
Abstract
Naringenin (NRG), as a flavanone from flavonoids family, is widely found in grapefruit, lemon tomato, and Citrus fruits. NRG has shown strong anti-inflammatory and antioxidant activities in body organs via mechanisms such as enhancement of glutathione S-transferase (GST), superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), and catalase (CAT) activity, but reduction of serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), lactate dehydrogenase (LDH), and malondialdehyde (MDA). Furthermore, NRG anti-apoptotic potential was indicated to be mediated by regulating B-cell lymphoma (Bcl-2), Bcl-2-associated X protein (Bax) and caspase3/9. Overall, these properties make NRG a highly fascinating compound with beneficial pharmacological effects. Based on the literature, NRG-induced protective effects against toxicities produced by natural toxins, pharmaceuticals, heavy metals, and environmental chemicals, were mainly mediated via suppression of lipid peroxidation, oxidative stress (through boosting the antioxidant arsenal), and inflammatory factors (e.g., TNF-α, interleukin [IL]-6, IL-10, and IL-12), and activation of PI3K/Akt and MAPK survival signaling pathways. Despite considerable body of evidence on protective properties of NRG against a variety of toxic compounds, more well-designed experimental studies and particularly, clinical trials are required before reaching a concrete conclusion. The present review discusses how NRG protects against the above-noted toxic compounds.
Collapse
Affiliation(s)
- Karim Naraki
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ramin Rezaee
- Clinical Research Unit, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Pharmaceutical Research Center Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
18
|
Pontifex MG, Malik MMAH, Connell E, Müller M, Vauzour D. Citrus Polyphenols in Brain Health and Disease: Current Perspectives. Front Neurosci 2021; 15:640648. [PMID: 33679318 PMCID: PMC7933480 DOI: 10.3389/fnins.2021.640648] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 01/25/2021] [Indexed: 12/17/2022] Open
Abstract
In addition to essential micronutrients such as vitamin C, citrus fruits represent a considerably rich source of non-essential bioactive compounds, in particular flavanones which form a sub-set of the flavonoid group. Preclinical studies have demonstrated the neuroprotective potential of citrus flavonoids and have highlighted both the well-established (anti-inflammatory and anti-oxidative properties), and newly emerging (influence upon blood-brain barrier function/integrity) mechanistic actions by which these neurological effects are mediated. Encouragingly, results from human studies, although limited in number, appear to support this preclinical basis, with improvements in cognitive performance and disease risk observed across healthy and disease states. Therefore, citrus fruits - both as whole fruit and 100% juices - should be encouraged within the diet for their potential neurological benefit. In addition, there should be further exploration of citrus polyphenols to establish therapeutic efficacy, particularly in the context of well-designed human interventions.
Collapse
Affiliation(s)
- Matthew G Pontifex
- Norwich Medical School, Biomedical Research Centre, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, United Kingdom
| | - Mohammad M A H Malik
- Norwich Medical School, Biomedical Research Centre, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, United Kingdom
| | - Emily Connell
- Norwich Medical School, Biomedical Research Centre, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, United Kingdom
| | - Michael Müller
- Norwich Medical School, Biomedical Research Centre, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, United Kingdom
| | - David Vauzour
- Norwich Medical School, Biomedical Research Centre, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, United Kingdom
| |
Collapse
|
19
|
Wang Y, Liu XJ, Chen JB, Cao JP, Li X, Sun CD. Citrus flavonoids and their antioxidant evaluation. Crit Rev Food Sci Nutr 2021; 62:3833-3854. [PMID: 33435726 DOI: 10.1080/10408398.2020.1870035] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The antioxidant ability is the link and bridge connecting a variety of biological activities. Citrus flavonoids play an essential role in regulating oxidative stress and are an important source of daily intake of antioxidant supplements. Many studies have shown that citrus flavonoids promote health through antioxidation. In this review, the biosynthesis, composition and distribution of citrus flavonoids were concluded. The detection methods of antioxidant capacity of citrus flavonoids were divided into four categories: chemical, cellular, animal and clinical antioxidant capacity evaluation systems. The modeling methods, applicable scenarios, and their relative merits were compared based on these four systems. The antioxidant functions of citrus flavonoids under different evaluation systems were also discussed, especially the regulation of the Nrf2-antioxidases pathway. Some shortcomings in the current research were pointed out, and some suggestions for progress were put forward.
Collapse
Affiliation(s)
- Yue Wang
- Laboratory of Fruit Quality Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Zhejiang University, Hangzhou, China
| | - Xiao-Juan Liu
- Laboratory of Fruit Quality Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Zhejiang University, Hangzhou, China
| | - Jie-Biao Chen
- Laboratory of Fruit Quality Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Zhejiang University, Hangzhou, China
| | - Jin-Ping Cao
- Laboratory of Fruit Quality Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Zhejiang University, Hangzhou, China
| | - Xian Li
- Laboratory of Fruit Quality Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Zhejiang University, Hangzhou, China
| | - Chong-De Sun
- Laboratory of Fruit Quality Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Zhejiang University, Hangzhou, China
| |
Collapse
|
20
|
Kesh S, Kannan RR, Balakrishnan A. Naringenin alleviates 6-hydroxydopamine induced Parkinsonism in SHSY5Y cells and zebrafish model. Comp Biochem Physiol C Toxicol Pharmacol 2021; 239:108893. [PMID: 32949818 DOI: 10.1016/j.cbpc.2020.108893] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 09/01/2020] [Accepted: 09/09/2020] [Indexed: 01/05/2023]
Abstract
6-Hydroxydopamine (6-OHDA) is a neurotoxin that inhibits the mitochondrial complex I causing mitochondrial impairment, aetiology of Parkinson's. Naringenin is a flavanone predominantly present in citrus fruits. Due to its high antioxidant and anti-inflammatory potential, it has been widely studied against various disorders. In this study, the neuroprotective effect of naringenin was determined against 6-OHDA induced toxicity with Levodopa (l-DOPA) as the standard. Naringenin reduced 6-OHDA induced oxidative stress biomarker levels such as CAT, GSH, SOD, and ROS. Naringenin rescued 6-OHDA induced reduction of the mitochondrial membrane potential. Treatment with naringenin improved the locomotion of the 6-OHDA treated zebrafish larvae which showed stagnant swimming patterns. Naringenin was also found to downregulate the expression of some Parkinsonian genes such as casp9, lrrk2, and polg and upregulate pink1. These studies attribute to naringenin as a viable molecule to study further for its neuroprotective effects against 6-OHDA induced neurotoxicity and neurodegeneration.
Collapse
Affiliation(s)
- Swathi Kesh
- Neuroscience Lab, Centre for Molecular and Nanomedical Sciences, Centre for Nanoscience and Nanotechnology, School of Bio and Chemical Engineering, Sathyabama Institute of Science and Technology, Chennai - 600119, Tamil Nadu, India
| | - Rajaretinam Rajesh Kannan
- Neuroscience Lab, Centre for Molecular and Nanomedical Sciences, Centre for Nanoscience and Nanotechnology, School of Bio and Chemical Engineering, Sathyabama Institute of Science and Technology, Chennai - 600119, Tamil Nadu, India..
| | - Anandan Balakrishnan
- Department of Genetics, University of Madras, Dr ALM PGIBMS Campus, Taramani, Chennai, Tamil Nadu, India
| |
Collapse
|
21
|
Arruda HS, Neri-Numa IA, Kido LA, Maróstica Júnior MR, Pastore GM. Recent advances and possibilities for the use of plant phenolic compounds to manage ageing-related diseases. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104203] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
22
|
Remedying the Mitochondria to Cure Human Diseases by Natural Products. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5232614. [PMID: 32733635 PMCID: PMC7376439 DOI: 10.1155/2020/5232614] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 06/05/2020] [Accepted: 06/25/2020] [Indexed: 12/15/2022]
Abstract
Mitochondria are the ‘engine' of cells. Mitochondrial dysfunction is an important mechanism in many human diseases. Many natural products could remedy the mitochondria to alleviate mitochondria-involved diseases. In this review, we summarized the current knowledge of the relationship between the mitochondria and human diseases and the regulation of natural products to the mitochondria. We proposed that the development of mitochondrial regulators/nutrients from natural products to remedy mitochondrial dysfunction represents an attractive strategy for a mitochondria-involved disorder therapy. Moreover, investigating the mitochondrial regulation of natural products can potentiate the in-depth comprehension of the mechanism of action of natural products.
Collapse
|
23
|
Zhou T, Liu L, Wang Q, Gao Y. Naringenin alleviates cognition deficits in high-fat diet-fed SAMP8 mice. J Food Biochem 2020; 44:e13375. [PMID: 32677738 DOI: 10.1111/jfbc.13375] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 06/06/2020] [Accepted: 06/19/2020] [Indexed: 01/03/2023]
Abstract
Naringenin is a natural dihydro flavonoid that is abundant in grapefruit. Previous studies suggested the cognition protective effect of naringenin in various cognitive deficits models, such as type 2 diabetic rat model and chemicals (e.g., lipopolysaccharide, scopolamine) treated rodents. However, the effects of naringenin on aging animals and the potential mechanisms are still unclear. In this study, we investigated the influence of naringenin administration on learning deficits in aging mice. High-fat diet-fed SAMP8 mice were employed as an age-related model of Alzheimer's disease. Dietary administration of 0.2% naringenin for 12 weeks significantly improved the spatial learning and memory performance of the high-fat diet-fed SAMP8 mice in both Barnes Maze test and Morris Water Maze test. Further mechanism research indicated that naringenin reduced Aβ production, tau-hyperphosphorylation, oxidative stress, and neuroinflammation in the brain. This research provides further evidence for the treatment effect of naringenin on Alzheimer's disease. PRACTICAL APPLICATIONS: Naringenin, also known as 4',5,7-thrihydroxyflflavanone, is a natural dihydro flavonoid that is abundant in grapefruit and other citrus fruits. The current study first demonstrated the improvement effect of naringenin on cognition deficits in HFD-fed SAMP8 mice, an aging mouse model. Potential mechanisms were also systematically explained by exploring the amyloid-β (Aβ) accumulation, tau hyperphosphorylation, oxidative stress, and neuroinflammation in the brain of mice. This study provides further evidence for the utilization of naringenin as an effective treatment agent for Alzheimer's disease.
Collapse
Affiliation(s)
- Tao Zhou
- Department of Neurosurgery, Brain Hospital, Weifang People's Hospital, Weifang, China
| | - Long Liu
- Department of Neurosurgery, Brain Hospital, Weifang People's Hospital, Weifang, China
| | - Qiulian Wang
- Department of Neurosurgery, Brain Hospital, Weifang People's Hospital, Weifang, China
| | - Ying Gao
- Department of Neurosurgery, Brain Hospital, Weifang People's Hospital, Weifang, China
| |
Collapse
|
24
|
UHPLC-HESI-OT-MS-MS Biomolecules Profiling, Antioxidant and Antibacterial Activity of the "Orange-Yellow Resin" from Zuccagnia punctata Cav. Antioxidants (Basel) 2020; 9:antiox9020123. [PMID: 32024138 PMCID: PMC7070614 DOI: 10.3390/antiox9020123] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/24/2020] [Accepted: 01/26/2020] [Indexed: 02/05/2023] Open
Abstract
This research was designed to investigate the metabolite profiling, phenolics, and flavonoids content as well as the potential antioxidant and antibacterial, properties of orange-yellow resin from Zuccagnia punctata Cav (ZpRe). Metabolite profiling was obtained by a ultrahigh resolution liquid chromatography orbitrap MS analysis (UHPLC-ESI-OT-MS-MS). The antioxidant properties were screened by four methods: 2,2-diphenyl-1-picrylhydrazyl assay (DPPH), trolox equivalent antioxidant activity assay (TEAC), ferric-reducing antioxidant power assay (FRAP), and lipid peroxidation in erythrocytes (LP)). The antibacterial activity was evaluated according to the Clinical and Laboratory Standards Institute (CLSI) rules. The resin displayed a strong DPPH scavenging activity (IC50 = 25.72 µg/mL) and showed a percentage of inhibition of LP close to that of the reference compound catechin (70% at 100 µg ZpRe/mL), while a moderated effect was observed in the FRAP and TEAC assays. The resin showed a content of phenolic and flavonoid compounds of 391 mg GAE/g and 313 mg EQ/g respectively. Fifty phenolics compounds were identified by ultrahigh resolution liquid chromatography orbitrap MS analysis (UHPLC-PDA-OT-MS) analysis. Thirty-one compounds are reported for the first time, updating the knowledge on the chemical profile of this species. The importance of the biomolecules identified support traditional use of this endemic plant. Furthermore, additional pharmacological data is presented that increase the potential interest of this plant for industrial sustainable applications.
Collapse
|
25
|
Nouri Z, Fakhri S, El-Senduny FF, Sanadgol N, Abd-ElGhani GE, Farzaei MH, Chen JT. On the Neuroprotective Effects of Naringenin: Pharmacological Targets, Signaling Pathways, Molecular Mechanisms, and Clinical Perspective. Biomolecules 2019; 9:E690. [PMID: 31684142 PMCID: PMC6920995 DOI: 10.3390/biom9110690] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 10/30/2019] [Accepted: 10/31/2019] [Indexed: 12/12/2022] Open
Abstract
As a group of progressive, chronic, and disabling disorders, neurodegenerative diseases (NDs) affect millions of people worldwide, and are on the rise. NDs are known as the gradual loss of neurons; however, their pathophysiological mechanisms have not been precisely revealed. Due to the complex pathophysiological mechanisms behind the neurodegeneration, investigating effective and multi-target treatments has remained a clinical challenge. Besides, appropriate neuroprotective agents are still lacking, which raises the need for new therapeutic agents. In recent years, several reports have introduced naturally-derived compounds as promising alternative treatments for NDs. Among natural entities, flavonoids are multi-target alternatives affecting different pathogenesis mechanisms in neurodegeneration. Naringenin is a natural flavonoid possessing neuroprotective activities. Increasing evidence has attained special attention on the variety of therapeutic targets along with complex signaling pathways for naringenin, which suggest its possible therapeutic applications in several NDs. Here, in this review, the neuroprotective effects of naringenin, as well as its related pharmacological targets, signaling pathways, molecular mechanisms, and clinical perspective, are described. Moreover, the need to develop novel naringenin delivery systems is also discussed to solve its widespread pharmacokinetic limitation.
Collapse
Affiliation(s)
- Zeinab Nouri
- Student's Research Committee, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah 6714415153, Iran.
| | - Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran.
| | - Fardous F El-Senduny
- Biochemistry division, Chemistry Department, Faculty of Science, Mansoura University, 35516 Mansoura, Egypt.
| | - Nima Sanadgol
- Department of Biology, Faculty of Sciences, University of Zabol, Zabol 7383198616, Iran.
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto 14040-903, Brazil.
| | - Ghada E Abd-ElGhani
- Department of Chemistry, Faculty of Science, University of Mansoura, 35516 Mansoura, Egypt.
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran.
| | - Jen-Tsung Chen
- Department of Life Sciences, National University of Kaohsiung, Kaohsiung 811, Taiwan.
| |
Collapse
|