1
|
İlhan İ, Ascı H, Buyukbayram Hİ, Imeci OB, Sevuk MA, Erol Z, Aksoy F, Milletsever A. The Impact of the High-Fructose Corn Syrup on Cardiac Damage via SIRT1/PGC1-α Pathway: Potential Ameliorative Effect of Selenium. Biol Trace Elem Res 2024; 202:5166-5176. [PMID: 38305829 PMCID: PMC11442503 DOI: 10.1007/s12011-024-04081-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/26/2024] [Indexed: 02/03/2024]
Abstract
High-fructose corn syrup (HFCS) has been a subject of intense debate due to its association with cardiovascular risks. This study investigates the potential protective effects of selenium (Se) supplementation against cardiac damage induced by HFCS. Thirty-two male Wistar albino rats were divided into four equal groups: control, CS (20%-HFCS), CS with Se (20%-HFCS, 0.3 mg/kg-Se), and Se (0.3 mg/kg-Se) only. After a 6-week period, heart and aorta tissues were collected for histopathological, immunohistochemical, biochemical, and genetic analyses. HFCS consumption led to severe cardiac pathologies, increased oxidative stress, and altered gene expressions associated with inflammation, apoptosis, and antioxidant defenses. In the CS group, pronounced oxidative stress within the cardiac tissue was concomitant with elevated Bcl-2-associated X protein (Bax) expression and diminished expressions of B-cell-lymphoma-2 (Bcl-2), nuclear factor erythroid 2-related factor 2 (Nrf2), peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1-α), and silenced information regulator 1 (SIRT1). Se supplementation mitigated these effects, showing protective properties. Immunohistochemical analysis supported these findings, demonstrating decreased expressions of caspase-3, tumor necrosis factor-alpha (TNF-α), IL-1β, and vascular endothelial growth factor (VEGF) in the CS + Se group compared to the CS group. The study suggests that Se supplementation exerts anti-inflammatory, antioxidant, and antiapoptotic effects, potentially attenuating HFCS-induced cardiovascular toxicity. These findings highlight the importance of dietary considerations and selenium supplementation in mitigating cardiovascular risks associated with HFCS consumption.
Collapse
Affiliation(s)
- İlter İlhan
- Faculty of Medicine, Department of Biochemistry, Suleyman Demirel University, Isparta, Turkey.
| | - Halil Ascı
- Faculty of Medicine, Department of Pharmacology, Suleyman Demirel University, Isparta, Turkey
| | | | - Orhan Berk Imeci
- Faculty of Medicine, Department of Pharmacology, Suleyman Demirel University, Isparta, Turkey
| | - Mehmet Abdulkadir Sevuk
- Faculty of Medicine, Department of Pharmacology, Suleyman Demirel University, Isparta, Turkey
| | - Zeki Erol
- Faculty of Veterinary, Department of Food Hygiene and Technology, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | - Fatih Aksoy
- Faculty of Medicine, Department of Pharmacology, Suleyman Demirel University, Isparta, Turkey
- Faculty of Medicine, Department of Cardiology, Suleyman Demirel University, Isparta, Turkey
| | - Adem Milletsever
- Faculty of Veterinary, Department of Pathology, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| |
Collapse
|
2
|
Tepebaşı MY, Savran M, Coşan S, Taştan ŞA, Aydın B. The protective role of selenium against high-fructose corn syrup-induced kidney damage: a histopathological and molecular analysis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7829-7837. [PMID: 38734838 PMCID: PMC11450133 DOI: 10.1007/s00210-024-03149-w] [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: 02/26/2024] [Accepted: 05/05/2024] [Indexed: 05/13/2024]
Abstract
With the growth of the food industry, fructose, the intake of which increases with food, causes obesity and metabolic syndrome. Kidney damage may develop from metabolic syndrome. Selenium (Se) participates in the structure of antioxidant enzymes and has a medicinal effect. In this work, the protective impact of Se on kidney damage produced by high-fructose corn syrup (HFCS) via endoplasmic reticulum (ER) stress was examined. The study comprised four groups, each consisting of ten experimental animals: control, HFCS (20%-HFCS), HFCS (20%-HFCS), + Se (0.3 mg/kg/day/po), and Se (0.3 mg/kg/day/po) alone. The duration of the experiment was 6 weeks. Kidney tissues were stained with hematoxylin and eosin for histological examination. Immunohistochemical analysis was conducted to assess TNF-α and caspase-3 levels. The spectrophotometric evaluation was performed to measure TOS (total oxidant status), TAS (total antioxidant status), and OSI (oxidative stress index) levels. The PERK, ATF4, CHOP, BCL-2, and caspase-9 gene expression levels were assessed by the RT-qPCR method. After Se treatment, histopathological abnormalities and TNF-α and caspase-3 levels in the HFCS+Se group decreased (p < 0.001). While TOS and OSI levels increased dramatically in the HFCS group, TAS values decreased significantly but improved after Se application (p < 0.001). The expression levels of the genes PERK, ATF4, CHOP, and caspase-9 were significantly lower in the HFCS group when compared to the HFCS+Se group (p < 0.05). Our findings suggest that Se may protect against ER stress, oxidative stress, apoptosis, and kidney damage caused by high-dose fructose consumption.
Collapse
Affiliation(s)
| | - Mehtap Savran
- Department of Medical Pharmacology, University of Süleyman Demirel, Isparta, Turkey
| | - Samet Coşan
- Department of Medical Pharmacology, University of Süleyman Demirel, Isparta, Turkey
| | | | - Bünyamin Aydın
- Department of Internal Medicine, Kütahya University of Health Sciences, Kütahya, Turkey
| |
Collapse
|
3
|
Lin S, Chen C, Ouyang P, Cai Z, Liu X, Abdurahman A, Peng J, Li Y, Zhang Z, Song GL. SELENOM Knockout Induces Synaptic Deficits and Cognitive Dysfunction by Influencing Brain Glucose Metabolism. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:1607-1619. [PMID: 36635091 DOI: 10.1021/acs.jafc.2c07491] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Selenium, a trace element associated with memory impairment and glucose metabolism, mainly exerts its function through selenoproteins. SELENOM is a selenoprotein located in the endoplasmic reticulum (ER) lumen. Our study demonstrates for the first time that SELENOM knockout decreases synaptic plasticity and causes memory impairment in 10-month-old mice. In addition, SELENOM knockout causes hyperglycaemia and disturbs glucose metabolism, which is essential for synapse formation and transmission in the brain. Further research reveals that SELENOM knockout leads to inhibition of the brain insulin signaling pathway [phosphatidylinositol 3-kinase (PI3K)/AKT/mTOR/p70 S6 kinase pathway], which may impair synaptic plasticity in mice. High-fat diet (HFD) feeding suppresses the brain insulin signaling pathway in SELENOM knockout mice and leads to earlier onset of cognitive impairment at 5 months of age. In general, our study demonstrates that SELENOM knockout induces synaptic deficits via the brain insulin signaling pathway, thus leading to cognitive dysfunction in mice. These data strongly suggest that SELENOM plays a vital role in brain glucose metabolism and contributes substantially to synaptic plasticity.
Collapse
Affiliation(s)
- Shujing Lin
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong518060, People's Republic of China
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, Guangdong518000, People's Republic of China
| | - Chen Chen
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong518060, People's Republic of China
| | - Pei Ouyang
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong518060, People's Republic of China
| | - Zhiyu Cai
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong518060, People's Republic of China
| | - Xibei Liu
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong518060, People's Republic of China
| | - Anwar Abdurahman
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong518060, People's Republic of China
| | - Jiaying Peng
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong518060, People's Republic of China
| | - Yu Li
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong518060, People's Republic of China
| | - Zhonghao Zhang
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong518060, People's Republic of China
| | - Guo-Li Song
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong518060, People's Republic of China
- Shenzhen Bay Laboratory, Shenzhen, Guangdong518000, People's Republic of China
| |
Collapse
|
4
|
Albrahim T. Lycopene Modulates Oxidative Stress and Inflammation in Hypercholesterolemic Rats. Pharmaceuticals (Basel) 2022; 15:1420. [PMID: 36422550 PMCID: PMC9693203 DOI: 10.3390/ph15111420] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 07/30/2023] Open
Abstract
The complicated disorder of hypercholesterolemia has several underlying factors, including genetic and lifestyle factors. Low LDL cholesterol and elevated serum total cholesterol are its defining features. A carotenoid with antioxidant quality is lycopene. Examining lycopene activity in an animal model of hypercholesterolemia induced using food was the aim of this investigation. Triglycerides, LDL cholesterol, HDL cholesterol, and plasma total cholesterol were all measured. Biomarkers of renal and cardiac function were also examined. Apoptotic indicators, pro-inflammatory markers, and oxidative stress were also assessed. Additionally, the mRNA expression of paraoxonase 1 (PON-1), peroxisome proliferator-activated receptor gamma (PPAR-γ), and PPAR-γ coactivator 1 alpha (PGC-1α) in cardiac and renal tissues was examined. Rats showed elevated serum lipid levels, renal and cardiac dysfunction, significant oxidative stress, and pro-inflammatory and apoptotic markers at the end of the study. Treatment with lycopene significantly corrected and restored these changes. Additionally, lycopene markedly increased the mRNA expression of PGC-1α and PON-1, and decreased PPAR-γ expression. It was determined that lycopene has the capacity to modulate the PPAR-γ and PON-1 signaling pathway in order to preserve the cellular energy metabolism of the heart and kidney, which in turn reduces tissue inflammatory response and apoptosis. According to these findings, lycopene may be utilized as a medication to treat hypercholesterolemia. However, further studies should be conducted first to determine the appropriate dose and any adverse effects that may appear after lycopene usage in humans.
Collapse
Affiliation(s)
- Tarfa Albrahim
- Department of Health Sciences, Clinical Nutrition, College of Health and Rehabilitation Sciences, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| |
Collapse
|
5
|
Restricted cafeteria feeding and treadmill exercise improved body composition, metabolic profile and exploratory behavior in obese male rats. Sci Rep 2022; 12:19545. [PMID: 36379981 PMCID: PMC9666649 DOI: 10.1038/s41598-022-23464-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/31/2022] [Indexed: 11/16/2022] Open
Abstract
The aim of this study was to evaluate, in male Long-Evans rats, whether a restricted-cafeteria diet (CAFR), based on a 30% calorie restriction vs continuous ad libitum cafeteria (CAF) fed animals, administered alone or in combination with moderate treadmill exercise (12 m/min, 35 min, 5 days/week for 8 weeks), was able to ameliorate obesity and the associated risk factors induced by CAF feeding for 18 weeks and to examine the changes in circadian locomotor activity, hypothalamic-pituitary-adrenal (HPA) axis functionality, and stress response elicited by this dietary pattern. In addition to the expected increase in body weight and adiposity, and the development of metabolic dysregulations compatible with Metabolic Syndrome, CAF intake resulted in a sedentary profile assessed by the home-cage activity test, reduced baseline HPA axis activity through decreased corticosterone levels, and boosted exploratory behavior. Both CAFR alone and in combination with exercise reduced abdominal adiposity and hypercholesterolemia compared to CAF. Exercise increased baseline locomotor activity in the home-cage in all dietary groups, boosted exploratory behavior in STD and CAF, partially decreased anxiety-like behavior in CAF and CAFR, but did not affect HPA axis-related parameters.
Collapse
|
6
|
Wu M, Zhai Y, Liang X, Chen W, Lin R, Ma L, Huang Y, Zhao D, Liang Y, Zhao W, Fang J, Fang S, Chen Y, Wang Q, Li W. Connecting the Dots Between Hypercholesterolemia and Alzheimer’s Disease: A Potential Mechanism Based on 27-Hydroxycholesterol. Front Neurosci 2022; 16:842814. [PMID: 35464321 PMCID: PMC9021879 DOI: 10.3389/fnins.2022.842814] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 03/01/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer’s disease (AD), the most common cause of dementia, is a complex and multifactorial disease involving genetic and environmental factors, with hypercholesterolemia considered as one of the risk factors. Numerous epidemiological studies have reported a positive association between AD and serum cholesterol levels, and experimental studies also provide evidence that elevated cholesterol levels accelerate AD pathology. However, the underlying mechanism of hypercholesterolemia accelerating AD pathogenesis is not clear. Here, we review the metabolism of cholesterol in the brain and focus on the role of oxysterols, aiming to reveal the link between hypercholesterolemia and AD. 27-hydroxycholesterol (27-OHC) is the major peripheral oxysterol that flows into the brain, and it affects β-amyloid (Aβ) production and elimination as well as influencing other pathogenic mechanisms of AD. Although the potential link between hypercholesterolemia and AD is well established, cholesterol-lowering drugs show mixed results in improving cognitive function. Nevertheless, drugs that target cholesterol exocytosis and conversion show benefits in improving AD pathology. Herbs and natural compounds with cholesterol-lowering properties also have a potential role in ameliorating cognition. Collectively, hypercholesterolemia is a causative risk factor for AD, and 27-OHC is likely a potential mechanism for hypercholesterolemia to promote AD pathology. Drugs that regulate cholesterol metabolism are probably beneficial for AD, but more research is needed to unravel the mechanisms involved in 27-OHC, which may lead to new therapeutic strategies for AD.
Collapse
Affiliation(s)
- Mingan Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yingying Zhai
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoyi Liang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weichun Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ruiyi Lin
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Linlin Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yi Huang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Di Zhao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yong Liang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Zhao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiansong Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shuhuan Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yunbo Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Qi Wang,
| | - Weirong Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
- Weirong Li,
| |
Collapse
|
7
|
Li LX, Chu JH, Chen XW, Gao PC, Wang ZY, Liu C, Fan RF. Selenium ameliorates mercuric chloride-induced brain damage through activating BDNF/TrKB/PI3K/AKT and inhibiting NF-κB signaling pathways. J Inorg Biochem 2022; 229:111716. [DOI: 10.1016/j.jinorgbio.2022.111716] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/27/2021] [Accepted: 01/01/2022] [Indexed: 02/07/2023]
|
8
|
Al Omairi NE, Albrakati A, Alsharif KF, Almalki AS, Alsanie W, Abd Elmageed ZY, Zaafar D, Lokman MS, Bauomy AA, Belal SK, Abdel-Daim MM, Abdel Moneim AE, Alyami H, Kassab RB. Selenium Nanoparticles with Prodigiosin Rescue Hippocampal Damage Associated with Epileptic Seizures Induced by Pentylenetetrazole in Rats. BIOLOGY 2022; 11:biology11030354. [PMID: 35336729 PMCID: PMC8945383 DOI: 10.3390/biology11030354] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/31/2022] [Accepted: 02/13/2022] [Indexed: 12/11/2022]
Abstract
Simple Summary Epilepsy is a chronic neurological disease characterized by neuronal hyper electrical activity and the development of unprovoked seizures. Although several antiepileptic drugs are currently available, their application is associated with undesirable adverse effects. In an attempt to find a novel antiepileptic medication with minimum side effects, we have investigated the potential neuroprotective activity of prodigiosin, a red pigment produced by bacterial species that have important pharmaceutical and biological activities biosynthesized with selenium formulation (SeNPs-PDG) against a murine epileptic model mediated by pentylenetetrazole. The main recorded findings revealed that SeNPs-PDG delayed the onset of epileptic seizures and decreased their duration significantly. Additionally, SeNPs-PDG prevented hippocampal cell loss, oxidative stress, neuroinflammation, restored the balance between excitatory and inhibitory neurotransmitters, and notably normalized the monoaminergic and cholinergic transmission. These promising findings indicate that SeNPs-PDG might serve as a naturally derived anticonvulsant agent due to their active antioxidant, anti-inflammatory, anti-apoptotic, and neuromodulatory properties. Abstract Background: Prodigiosin (PDG) is a red pigment synthesized by bacterial species with important pharmaceutical and biological activities. Here, we investigated the neuroprotective and anticonvulsant activities of green biosynthesized selenium formulations with PDG (SeNPs-PDG) versus pentylenetetrazole (PTZ)-induced epileptic seizures. Methods: Rats were assigned into six experimental groups: control; PTZ (60 mg/kg, epileptic model); sodium valproate (200 mg/kg) + PTZ; PDG (300 mg/kg) + PTZ; sodium selenite (0.5 mg/kg) + PTZ; and SeNPs-PDG (0.5 mg/kg) + PTZ. The treatment duration is extended to 28 days. Results: SeNPs-PDG pre-treatment delayed seizures onset and reduced duration upon PTZ injection. Additionally, SeNPs-PDG enhanced the antioxidant capacity of hippocampal tissue by activating the expression of nuclear factor erythroid 2–related factor 2 and innate antioxidants (glutathione and glutathione derivatives, in addition to superoxide dismutase and catalase) and decreasing the levels of pro-oxidants (lipoperoxidation products and nitric oxide). SeNPs-PDG administration inhibited inflammatory reactions associated with epileptic seizure development by suppressing the production and activity of glial fibrillary acidic protein and pro-inflammatory mediators, including interleukin-1 beta, tumor necrosis factor-alpha, cyclooxygenase-2, inducible nitric oxide synthase, and nuclear factor kappa B. Moreover, SeNPs-PDG protected against hippocampal cell loss following PTZ injection by decreasing the levels of cytosolic cytochrome c, Bax, and caspase-3 and enhancing the expression of anti-apoptotic Bcl-2. Interestingly, SeNPs-PDG restored the PTZ-induced imbalance between excitatory and inhibitory amino acids and improved monoaminergic and cholinergic transmission. Conclusions: These promising antioxidative, anti-inflammatory, anti-apoptotic, and neuromodulatory activities indicate that SeNPs-PDG might serve as a naturally derived anticonvulsant agent.
Collapse
Affiliation(s)
- Naif E. Al Omairi
- Department of Internal Medicine, College of Medicine, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (N.E.A.O.); (H.A.)
| | - Ashraf Albrakati
- Department of Human Anatomy, College of Medicine, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
- Correspondence: ; Tel.: +966-555696608
| | - Khalaf F. Alsharif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (K.F.A.); (W.A.)
| | | | - Walaa Alsanie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (K.F.A.); (W.A.)
| | - Zakaria Y. Abd Elmageed
- Department of Pharmacology, Edward Via College of Osteopathic Medicine, University of Louisiana at Monroe, Monroe, LA 71203, USA;
| | - Dalia Zaafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology and Information, Cairo 11311, Egypt;
| | - Maha S. Lokman
- Biology Department, College of Science and Humanities, Prince Sattam bin Abdul Aziz University, Alkharj 11942, Saudi Arabia;
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo 11795, Egypt; (A.E.A.M.); (R.B.K.)
| | - Amira A. Bauomy
- Department of Science Laboratories, College of Science and Arts, Qassim University, ArRass 52719, Saudi Arabia;
| | - Saied K. Belal
- Department of Human Anatomy, College of Medicine, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Mohamed M. Abdel-Daim
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia;
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Ahmed E. Abdel Moneim
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo 11795, Egypt; (A.E.A.M.); (R.B.K.)
| | - Hussain Alyami
- Department of Internal Medicine, College of Medicine, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (N.E.A.O.); (H.A.)
| | - Rami B. Kassab
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo 11795, Egypt; (A.E.A.M.); (R.B.K.)
- Biology Department, Faculty of Science and Arts, Al-Baha University, Al-Mukhwah 65554, Saudi Arabia
| |
Collapse
|
9
|
Soleimani Asl S, Amiri I, Samzadeh-Kermani A, Abbasalipourkabir R, Gholamigeravand B, Shahidi S. Chitosan-coated Selenium nanoparticles enhance the efficiency of stem cells in the neuroprotection of streptozotocin-induced neurotoxicity in male rats. Int J Biochem Cell Biol 2021; 141:106089. [PMID: 34601090 DOI: 10.1016/j.biocel.2021.106089] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/23/2021] [Accepted: 09/25/2021] [Indexed: 12/27/2022]
Abstract
Alzheimer's disease (AD) is one of the common neurodegenerative diseases characterized by memory impairment. The protective effects of stem cell-based therapy have been reported in AD. In this study, it was assumed that Chitosan-coated Selenium nanoparticles (ChSeNPs) increase the efficiency of stem cells in the attenuation of neurotoxicity in the rat AD model. The AD model was induced using Streptozotocin (STZ) and treated by the adipose-derived mesenchymal stem cells (AMSCs) and SeNPs/ChSeNPs (0.4 mg/kg). Passive avoidance learning and recognition memory were assessed using shuttle box and novel object recognition tasks. The amyloid-beta deposition, the injected cells' homing and survival, antioxidant capacity, and BDNF concentration were evaluated using the histological, biochemical, and ELISA methods. The results showed that the combined administration of ChSeNPs and AMSCs is more effective in increasing the step-through latency and discrimination index than administering SeNPs and stem cells. Combined therapy caused a significant increase in antioxidant capacity that ChSeNPs was more effective than SeNPs, while AMSCs beside SeNPs had a greater effect on BDNF levels compared to conventional treatment of nanoparticles or AMSCs alone. Ultimately, the homing and survival of the transplanted AMSCs were greater in the group that received both stem cells and ChSeNPs. Taken together, it seems that the administration of ChSeNPs enhances the efficiency of transplanted stem cells in decreasing the neurotoxicity induced by STZ through an increase in the antioxidant capacity.
Collapse
Affiliation(s)
- Sara Soleimani Asl
- Endometrium and Endometriosis Research Centre, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Anatomy, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Iraj Amiri
- Endometrium and Endometriosis Research Centre, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Anatomy, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Roghayeh Abbasalipourkabir
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Bahareh Gholamigeravand
- Department of Anatomy, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Siamak Shahidi
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
10
|
Gholamigeravand B, Shahidi S, Afshar S, Gholipour P, Samzadeh-Kermani A, Amiri K, Majidi M, Abbasalipourkabir R, Arabestani MR, Soleimani Asl S. Synergistic effects of adipose-derived mesenchymal stem cells and selenium nanoparticles on streptozotocin-induced memory impairment in the rat. Life Sci 2021; 272:119246. [PMID: 33607156 DOI: 10.1016/j.lfs.2021.119246] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/02/2021] [Accepted: 02/09/2021] [Indexed: 12/19/2022]
Abstract
AIMS Memory impairment is determined to be the most well-known symptom of Alzheimer's disease (AD). Although cell therapy seems is an efficient therapeutic strategy to attenuate the AD-related memory impairment, transplanted cells have a short lifespan and do not survive long term in the recipient animals. Herein, we investigated whether the combination therapy of Selenium nanoparticles (SeNPs) and stem cells attenuates the neurotoxicity in an AD animal model. MATERIAL AND METHODS The adipose-derived mesenchymal stem cells (AMSCs) were transplanted in the AD model. In addition to cell injections, the animals also received oral administration of SeNPs (0.4 mg/kg) for one month. Recognition memory, cell survival, and BDNF concentration were assessed using the novel object recognition task, immunofluorescence, and ELISA methods. KEY FINDINGS Our results showed that the combined therapy was more effective in increasing the discrimination index than the administering SeNPs or AMSCs alone. Moreover, SeNPs and stem cells together had the greatest effects in reducing the deposition of Aβ and increasing the concentration of BDNF. Ultimately, the survival and proliferation of transplanted cells were more in the group that received stem cells besides SeNPs. SIGNIFICANCE Taken together, it seems that the transplantation of MSCs combined with SeNPs could achieve better results in the neuroprotection in the AD model than a conventional treatment of SeNPs or stem cells alone.
Collapse
Affiliation(s)
- Bahareh Gholamigeravand
- Endometrium and Endometriosis Research Centre, Department of Anatomy, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Siamak Shahidi
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Simin Afshar
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Parsa Gholipour
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Kimia Amiri
- School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mahsa Majidi
- School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Mohammad Reza Arabestani
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sara Soleimani Asl
- Endometrium and Endometriosis Research Centre, Department of Anatomy, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
11
|
Ye T, Gao HW, Xuan WT, Ye S, Zhou P, Li XQ, Wang Y, Song H, Liu YY, Cai B. The Regulating Mechanism of Chrysophanol on Protein Level of CaM-CaMKIV to Protect PC12 Cells Against Aβ 25-35-Induced Damage. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:2715-2723. [PMID: 32764873 PMCID: PMC7381772 DOI: 10.2147/dddt.s245128] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 06/30/2020] [Indexed: 12/14/2022]
Abstract
Objective To investigate the neuroprotective effect of chrysophanol (CHR) on PC12 treated with Aβ25-35, and the involved mechanism. Methods After the establishment of an AD cell model induced by Aβ25-35, the cell survival rate was detected by MTT, cell apoptosis was assayed by Hoechst 33342 staining, mRNA expressions of calmodulin (CaM), calcium/calmodulin-dependent protein kinase kinase (CaMKK), calcium/calmodulin-dependent protein kinase IV (CaMKIV) and tau (MAPT; commonly known as tau) were determined by qRT-PCR, and protein levels of CaM, CaMKK, CaMKIV, phospho-CaMKIV (p-CaMKIV), tau and phospho-tau (p-tau) were detected by Western blot analysis. Results When pretreated with CHR before exposure to Aβ25-35, PC12 cells showed that increased cell viability and reduced apoptosis. The qRT-PCR results indicated that the deposition of Aβ25-35 triggers a decrease in levels of CaM, CaMKK, CaMKIV, and tau in PC12 cells. In addition, Western blot results also suggested that Aβ25-35 decreases the protein expression of CaM, CaMKK, CaMKIV, p-CaMKIV, and the ratio of p-tau to tau in PC12 cells. However, the above effects were significantly alleviated after the treatment of CHR. Conclusion CHR plays a neuroprotective role in AD though decreasing the protein level of CaM-CaMKK-CaMKIV and the expression of p-tau downstream.
Collapse
Affiliation(s)
- Ting Ye
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China
| | - Hua-Wu Gao
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China
| | - Wei-Ting Xuan
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China
| | - Shu Ye
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, People's Republic of China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, People's Republic of China
| | - Peng Zhou
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, People's Republic of China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, People's Republic of China
| | - Xin-Quan Li
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China
| | - Yan Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, People's Republic of China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, People's Republic of China
| | - Hang Song
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, People's Republic of China
| | - Yan-Yan Liu
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, People's Republic of China
| | - Biao Cai
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, People's Republic of China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei 230012, People's Republic of China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, People's Republic of China
| |
Collapse
|
12
|
Yuan L, Qian L, Qian Y, Liu J, Yang K, Huang Y, Wang C, Li Y, Mu X. Bisphenol F-Induced Neurotoxicity toward Zebrafish Embryos. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2019; 53:14638-14648. [PMID: 31702913 DOI: 10.1021/acs.est.9b04097] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
In this study, the influence of bisphenol F (BPF) toward central nervous system (CNS) was assessed using zebrafish embryos. We found that BPF could induce significant neurotoxicity toward zebrafish embryos, including inhibited locomotion, reduced moving distance, and CNS cell apoptosis at an effective concentration of 0.0005 mg/L. Immunofluorescence assay showed that both microglia and astrocyte in zebrafish brain were significantly activated by BPF, indicating the existence of neuroinflammatory response. Peripheral motor neuron development was significantly inhibited by BPF at 72 hpf. RNA-seq data indicated that neuronal developmental processes and cell apoptosis pathways were significantly affected by BPF exposure, which was consistent with the phenotypic results. Chip-seq assay implied that the transcriptional changes were not mediated by ERα. Additionally, no significant change was found in neurotransmitter levels (5-hydroxytryptamine, dopamine, and acetylcholine) or acetylcholinesterase (Ache) enzyme activity after BPF exposure, indicating that BPF may not affect neurotransmission. In conclusion, BPF could lead to abnormal neural outcomes during zebrafish early life stage through inducing neuroinflammation and CNS cell apoptosis even at environmentally relevant concentration.
Collapse
Affiliation(s)
- Lilai Yuan
- Fishery Resource and Environment Research Center , Chinese Academy of Fishery Sciences , Beijing 100141 , People's Republic of China
| | - Le Qian
- College of Sciences , China Agricultural University , Beijing 100193 , People's Republic of China
| | - Yu Qian
- Fishery Resource and Environment Research Center , Chinese Academy of Fishery Sciences , Beijing 100141 , People's Republic of China
| | - Jia Liu
- Fishery Resource and Environment Research Center , Chinese Academy of Fishery Sciences , Beijing 100141 , People's Republic of China
| | - Ke Yang
- Fishery Resource and Environment Research Center , Chinese Academy of Fishery Sciences , Beijing 100141 , People's Republic of China
| | - Ying Huang
- Fishery Resource and Environment Research Center , Chinese Academy of Fishery Sciences , Beijing 100141 , People's Republic of China
| | - Chengju Wang
- College of Sciences , China Agricultural University , Beijing 100193 , People's Republic of China
| | - Yingren Li
- Fishery Resource and Environment Research Center , Chinese Academy of Fishery Sciences , Beijing 100141 , People's Republic of China
| | - Xiyan Mu
- Fishery Resource and Environment Research Center , Chinese Academy of Fishery Sciences , Beijing 100141 , People's Republic of China
| |
Collapse
|
13
|
Li B, Mo L, Yang Y, Zhang S, Xu J, Ge Y, Xu Y, Shi Y, Le G. Processing milk causes the formation of protein oxidation products which impair spatial learning and memory in rats. RSC Adv 2019; 9:22161-22175. [PMID: 35519476 PMCID: PMC9066704 DOI: 10.1039/c9ra03223a] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/08/2019] [Indexed: 12/20/2022] Open
Abstract
This study explored the effects of protein oxidation during milk processing on spatial learning and memory in rats. Increasing the heating time, fat content, and inlet air temperature during processing by boiling, microwave heating, spray-drying, or freeze-drying increases milk protein oxidation. Oxidative damage done to milk proteins by microwave heating is greater than that caused by boiling. Dityrosine (DT), as a kind of tyrosine oxidation product, is the most important marker of this process, especially during spray-drying. Rats received diets containing either SWM (spray-dried milk powder diet), FWM (freeze-dried milk powder diet), FWM + LDT (freeze-dried milk powder + low dityrosine diet, DT: 1.4 mg kg-1), or FWM + HDT (freeze-dried milk powder + high dityrosine diet, DT: 2.8 mg kg-1) for 6 weeks. We found that the SWM group, the FWM + LDT group, and the FWM + HDT group appeared to have various degrees of redox state imbalance and oxidative damage in plasma, liver, and brain tissues. Further, hippocampal inflammatory and apoptosis genes were significantly up-regulated in such groups, while learning and memory genes were significantly down-regulated. Eventually, varying degrees of spatial learning and memory impairment were demonstrated in those groups in the Morris water maze. This means that humans should control milk protein oxidation and improve the processing methods applied to food.
Collapse
Affiliation(s)
- Bowen Li
- The State Key Laboratory of Food Science and Technology, Jiangnan University Li Hu Avenue 1800 Wuxi PR China 214122 +86 510 85917789 +86 510 85869236 +86 510 85917789 +86 13812519691
- Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University Wuxi PR China 214122
| | - Ling Mo
- Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University Wuxi PR China 214122
- School of Public Health, Guilin Medical University Guilin PR China 541001
| | - Yuhui Yang
- Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University Wuxi PR China 214122
- College of Grain and Food Science, Henan University of Technology Zhengzhou PR China 450001
| | - Shuai Zhang
- Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University Wuxi PR China 214122
| | - Jingbing Xu
- Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University Wuxi PR China 214122
| | - Yueting Ge
- Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University Wuxi PR China 214122
| | - Yuncong Xu
- Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University Wuxi PR China 214122
| | - Yonghui Shi
- The State Key Laboratory of Food Science and Technology, Jiangnan University Li Hu Avenue 1800 Wuxi PR China 214122 +86 510 85917789 +86 510 85869236 +86 510 85917789 +86 13812519691
- Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University Wuxi PR China 214122
| | - Guowei Le
- The State Key Laboratory of Food Science and Technology, Jiangnan University Li Hu Avenue 1800 Wuxi PR China 214122 +86 510 85917789 +86 510 85869236 +86 510 85917789 +86 13812519691
- Center for Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University Wuxi PR China 214122
| |
Collapse
|