1
|
Divvela SSK, Gallorini M, Gellisch M, Patel GD, Saso L, Brand-Saberi B. Navigating redox imbalance: the role of oxidative stress in embryonic development and long-term health outcomes. Front Cell Dev Biol 2025; 13:1521336. [PMID: 40206404 PMCID: PMC11979171 DOI: 10.3389/fcell.2025.1521336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/19/2025] [Indexed: 04/11/2025] Open
Abstract
Embryonic development is a complex process of concurrent events comprising cell proliferation, differentiation, morphogenesis, migration, and tissue remodeling. To cope with the demands arising from these developmental processes, cells increase their nutrient uptake, which subsequently increases their metabolic activity. Mitochondria play a key role in the maintenance of metabolism and production of reactive oxygen species (ROS) as a natural byproduct. Regulation of ROS by antioxidants is critical and tightly regulated during embryonic development, as dysregulation results in oxidative stress that damages essential cellular components such as DNA, proteins, and lipids, which are crucial for cellular maintenance and in extension development. However, during development, exposure to certain exogenous factors or damage to cellular components can result in an imbalance between ROS production and its neutralization by antioxidants, leading to detrimental effects on the developmental process. In this review article, we highlight the crucial role of redox homeostasis in normal development and how disruptions in redox balance may result in developmental defects.
Collapse
Affiliation(s)
| | - Marialucia Gallorini
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Morris Gellisch
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| | - Gaurav Deepak Patel
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
2
|
Khan AA, Dewald HD. Nitric oxide and peroxynitrite as new biomarkers for early diagnosis of autism. Brain Res 2025; 1850:149438. [PMID: 39793916 DOI: 10.1016/j.brainres.2024.149438] [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: 07/15/2024] [Revised: 12/05/2024] [Accepted: 12/28/2024] [Indexed: 01/13/2025]
Abstract
Autism spectrum disorder, or autism, is a neurodevelopmental disorder of the developing child's brain with a genetic causality. It can be diagnosed at about three years after birth when it begins to present itself via a range of neuropsychiatric symptoms. Nitric oxide is a crucial small molecule of life synthesized within cells of our body systems, including cells of our brain. Peroxynitrite is the product of reaction between superoxide anion and nitric oxide. It normally isomerizes into harmless nitrates or nitrites. However, when excessive superoxide anion is present, the cellular concentration of peroxynitrite can increase to a toxic level. Autism has been suggested to cause oxidative damage to brain cells. Until now, it is impossible to sample tissue from a live brain. Instead, stem cells can be derived (from an autism patient's somatic cells) which can then be differentiated and chemically directed to grow into miniature 3-dimensional tissue masses resembling specific brain regions (e.g., the cortex) called brain organoids. This review discusses utilizing nitric oxide and peroxynitrite as biomarkers and comparing their relative concentrations in stem cells and stem cell derived brain organoids of healthy and autistic individuals to develop a bioanalytical process for early diagnosis of autism.
Collapse
Affiliation(s)
- Abdullah Asif Khan
- Department of Chemistry and Biochemistry, Ohio University, Athens, OH, United States
| | - Howard D Dewald
- Department of Chemistry and Biochemistry, Ohio University, Athens, OH, United States.
| |
Collapse
|
3
|
Jia Y, Kong X, Li R, Wang H, Li C, Cheng S, Duan W, Xiao Y, Mai Y, Deng W, Liu Y. Enhanced nasal-to-brain drug delivery by multivalent bioadhesive nanoparticle clusters for cerebral ischemic reperfusion injury protection. Acta Biomater 2025; 194:411-427. [PMID: 39870153 DOI: 10.1016/j.actbio.2025.01.036] [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: 08/02/2024] [Revised: 01/17/2025] [Accepted: 01/22/2025] [Indexed: 01/29/2025]
Abstract
Following cerebral ischemia, reperfusion injury can worsen ischemia-induced functional, metabolic disturbances, and pathological damage upon blood flow restoration, potentially leading to irreversible harm. Yet, there's a dearth of advanced, localized drug delivery systems ensuring active pharmaceutical ingredient (API) efficacy in cerebral protection during ischemia-reperfusion. This study introduces a multivalent bioadhesive nanoparticle-cluster, merging bioadhesive nanoparticles (BNPs) with dendritic polyamidoamine (PAMAM), enhancing nose-to-brain delivery and brain protection efficacy against cerebral ischemia-reperfusion injuries (CIRI). The BNPs-PAMAM cluster exhibits superior adhesion within the rat nasal cavity, prolonged retention, enabling sustained drug release, cerebral transportation, and accumulation, resulting in enhanced intracerebral pharmacokinetic profile. Intranasal administration circumvents systemic delivery challenges, ensuring CIRI protection drugs reach ischemic areas pre-reperfusion, overcoming thrombus-related delays. Administering BNPs-PAMAM loaded with dexmedetomidine (DEX) pre-reperfusion effectively prevents neuron apoptosis by α2-adrenoceptor activation, modulating the ischemic microenvironment, exerting triple neuroprotective effects against cerebral reperfusion injury. Importantly, only therapeutic DEX releases and accumulates in the nasal cavity, averting brain nanomaterial toxicity, promising for repeat administrations. This study presents a translational platform for nasal-to-brain drug delivery in CNS disease treatment. STATEMENT OF SIGNIFICANCE: Innovative Drug Delivery System: This study introduces a multivalent bioadhesive nanoparticle-cluster (BNPs-PAMAM) to enhance nasal-to-brain drug delivery for cerebral ischemia-reperfusion injury (CIRI) treatment. Enhanced Retention and Efficacy: The BNPs-PAMAM system significantly improves drug retention in the nasal cavity and ensures sustained release, thereby enhancing the therapeutic efficacy of the neuroprotective agent dexmedetomidine (DEX). Blood-Brain Barrier Circumvention: By leveraging intranasal administration, the system bypasses the blood-brain barrier, delivering DEX directly to ischemic brain regions before reperfusion and minimizing systemic side effects. Triple Neuroprotective Effects for CIRI protection: DEX delivered via BNPs-PAMAM effectively reduces oxidative stress and inflammation while enhancing mitochondrial autophagy, providing comprehensive protection against neuronal damage.
Collapse
Affiliation(s)
- Yizhen Jia
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Xiaohan Kong
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Rui Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Han Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Chujie Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Shihong Cheng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Wei Duan
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yan Xiao
- Laboratory Animal Center, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yang Mai
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Wenbin Deng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China; School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| | - Yang Liu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China; School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510275, China.
| |
Collapse
|
4
|
Li Y, Yu M, Wei Y, Zhou Z, Guo Y, Yuan M, Jin J, Li J, Shen H, Wu D. Risk assessment of developmental and neurotoxicity by the flavoring agent perillaldehyde: NAC (N-acetylcysteine) mitigation of oxidative stress-mediated inhibition of the Nrf2 pathway. Comp Biochem Physiol C Toxicol Pharmacol 2025; 288:110071. [PMID: 39549861 DOI: 10.1016/j.cbpc.2024.110071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/08/2024] [Accepted: 11/07/2024] [Indexed: 11/18/2024]
Abstract
Perillaldehyde (PAE), a prevalent flavoring agent, has raised safety concerns due to conflicting evidence regarding its toxicity. This study provides a comprehensive assessment of the developmental and neurotoxic effects of PAE in zebrafish, elucidating the underlying mechanisms of its toxicity. Results showed that PAE affected the viability and hatching rate of zebrafish at 96 h postfertilization with the 50 % lethal concentration (LC50) of 7.975 mg/L. Furthermore, exposed to a non-lethal concentration of 4 mg/L PAE induced a spectrum of morphological abnormalities, such as pericardial edema, delayed yolk sac absorption, reduced body length, and microphthalmia. Behavioral observations revealed that PAE reduced motor ability, and was accompanied by an increase in spontaneous turning angle and angular velocity. Using the TG(elav13:EGFP) transgenic model, we observed the number of newborn neurons was reduced, indicating that PAE induced obvious neurotoxic effects. Additionally, this concentration facilitated the accumulation of reactive oxygen species (ROS) and malondialdehyde (MDA), concomitantly decreasing the activity of antioxidant enzymes. QRT-PCR analysis revealed that PAE down-regulated Nestin and Neurogenin1 gene expression, up-regulated Glipr1a and Nox1 gene expression, and inhibited the Nrf2/HO-1 pathway. Notably, co-administration of N-acetylcysteine (NAC), an inhibitor of oxidative stress, mitigated oxidative stress levels and partially ameliorated the neurotoxicity. These findings suggest that oxidative stress is the primary mediator of PAE-induced neurotoxicity. This study provides crucial insights for the safe application of PAE.
Collapse
Affiliation(s)
- Yue Li
- Key Laboratory of Microecology-immune Regulatory Network and Related Diseases School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang Province 154007, PR China. https://twitter.com/LiYueJMSU
| | - Manchun Yu
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang 154007, PR China
| | - Ying Wei
- Key Laboratory of Microecology-immune Regulatory Network and Related Diseases School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang Province 154007, PR China
| | - Zhuoshuo Zhou
- Key Laboratory of Microecology-immune Regulatory Network and Related Diseases School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang Province 154007, PR China
| | - Yingxue Guo
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang 154007, PR China
| | - Min Yuan
- Science and Technology Innovation Center for College Students, Jiamusi University, Jiamusi, Heilongjiang 154007, PR China
| | - Jiazheng Jin
- Science and Technology Innovation Center for College Students, Jiamusi University, Jiamusi, Heilongjiang 154007, PR China
| | - Jinlian Li
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang 154007, PR China.
| | - Hongkuan Shen
- Jiamusi Inspection and Testing Center, Jiamusi, Heilongjiang 154007, PR China.
| | - Dongmei Wu
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang 154007, PR China.
| |
Collapse
|
5
|
Guo Y, Wang Y, Xu B, Li Y. The prospective therapeutic benefits of sesamol: neuroprotection in neurological diseases. Nutr Neurosci 2025:1-14. [PMID: 39881218 DOI: 10.1080/1028415x.2025.2457051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Oxidative stress is recognized as a critical contributor to the advancement of neurological diseases, thereby rendering the alleviation of oxidative stress a pivotal strategy in the therapeutic management of such conditions. Sesamol, the principal constituent of sesame oil, has been the subject of extensive research due to its significant antioxidant properties, especially its ability to effectively counteract oxidative stress within the central nervous system and confer neuroprotection. While sesamol demonstrates potential in the treatment and prevention of neurological diseases, its modulation of oxidative stress is complex and not yet fully understood. This review delves into the neuroprotective effects arising from sesamol's antioxidant properties, analyzing how its antioxidative capabilities impact neurological diseases. It provides a theoretical foundation and unveils potential novel therapeutic applications of sesamol in the treatment of neurological disorders through the modulation of oxidative stress.
Collapse
Affiliation(s)
- Yuchao Guo
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Yaqing Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, People's Republic of China
| | - Boyang Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, People's Republic of China
| | - Yue Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, People's Republic of China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macau, People's Republic of China
| |
Collapse
|
6
|
Ma R, You H, Liu H, Bao J, Zhang M. Hesperidin:a citrus plant component, plays a role in the central nervous system. Heliyon 2024; 10:e38937. [PMID: 39553629 PMCID: PMC11564962 DOI: 10.1016/j.heliyon.2024.e38937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 09/22/2024] [Accepted: 10/02/2024] [Indexed: 11/19/2024] Open
Abstract
Hesperidin is a kind of flavonoids, which has the biological activities of antioxidation, anti-inflammation, antibacterial, anti-virus, anti-allergy, anti-cancer, heart protection and neuroprotection. More and more studies have begun to pay attention to the therapeutic prospect of hesperidin in central nervous system (CNS) diseases. This paper describes its current role in the treatment of central nervous system diseases, especially stroke, and discusses its bioavailability, so as to provide a theoretical basis for the clinical application of hesperidin.
Collapse
Affiliation(s)
- Rui Ma
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Hong You
- Sino-French Neurorehabilitation Department of Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Hong Liu
- Clinical Medical College of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Juan Bao
- Clinical Medical College of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Min Zhang
- Sino-French Neurorehabilitation Department of Gansu Provincial Hospital, Lanzhou, Gansu, China
| |
Collapse
|
7
|
Nie S, Zhang S, Wu R, Zhao Y, Wang Y, Wang X, Zhu M, Huang P. Scutellarin: pharmacological effects and therapeutic mechanisms in chronic diseases. Front Pharmacol 2024; 15:1470879. [PMID: 39575387 PMCID: PMC11578714 DOI: 10.3389/fphar.2024.1470879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 10/17/2024] [Indexed: 11/24/2024] Open
Abstract
Scutellarin (SCU), a flavonoid glucuronide derived from Scutellaria barbata and Erigeron breviscapus, exhibits broad pharmacological effects with promising therapeutic potential in treating various chronic diseases. It has demonstrated efficacy in modulating multiple biological pathways, including antioxidant, anti-inflammatory, anti-apoptotic, and vasodilatory mechanisms. These protective roles make SCU a valuable compound in treating chronic diseases such as cerebrovascular diseases, cardiovascular diseases, neurodegenerative disorders, and metabolic diseases. Despite its multi-targeted effects, SCU faces challenges such as low bioavailability and limited clinical data, which hinder its widespread therapeutic application. Current research supports its potential to prevent oxidative stress, reduce inflammatory responses, and enhance cell survival in cells and rats. However, more comprehensive studies are required to clarify its molecular mechanisms and to develop strategies that enhance its bioavailability for clinical use. SCU could emerge as a potent therapeutic agent for the treatment of chronic diseases with complex pathophysiological mechanisms. This review examines the current literature on Scutellarin to provide a comprehensive understanding of its pharmacological activity, mechanisms of action, and therapeutic potential in treating chronic diseases.
Collapse
Affiliation(s)
- Shanshan Nie
- Department of Cardiovascular Disease, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Shan Zhang
- Department of Digestive Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Ruipeng Wu
- Department of Cardiovascular Disease, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Yuhang Zhao
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yongxia Wang
- Department of Cardiovascular Disease, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xinlu Wang
- Department of Cardiovascular Disease, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Mingjun Zhu
- Department of Cardiovascular Disease, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Peng Huang
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
8
|
Li X, Zheng K, Chen H, Li W. Ginsenoside Re Regulates Oxidative Stress through the PI3K/Akt/Nrf2 Signaling Pathway in Mice with Scopolamine-Induced Memory Impairments. Curr Issues Mol Biol 2024; 46:11359-11374. [PMID: 39451557 PMCID: PMC11506191 DOI: 10.3390/cimb46100677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/05/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024] Open
Abstract
While Ginsenoside Re has been shown to protect the central nervous system, reports of its effects on memory in the model of scopolamine-induced memory impairment are rare. The aim of this study was to investigate the effects of Ginsenoside Re on scopolamine (SCOP)-induced memory damage and the mechanism of action. Male ICR mice were treated with SCOP (3 mg/kg) for 7 days and with or without Ginsenoside Re for 14 days. As evidenced by behavioral studies (escape latency and cross platform position), brain tissue morphology, and oxidative stress indicators after Ginsenoside Re treatment, the memory damage caused by SCOP was significantly ameliorated. Further mechanism research indicated that Ginsenoside Re inhibited cell apoptosis by regulating the PI3K/Akt/Nrf2 pathway, thereby exerting a cognitive impairment improvement effect. This research suggests that Ginsenoside Re could protect against SCOP-induced memory defects possibly through inhibiting oxidative stress and cell apoptosis.
Collapse
Affiliation(s)
- Xin Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | | | | | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| |
Collapse
|
9
|
Lemus-Varela L, Torres-Mendoza B, Rabago-Domingo P, Cárdenas-Bedoya J, Zúñiga-González GM, Torres-Sanchez ED, Gabriel-Ortiz G. Impact of Early- and High-Dose Caffeine on the Cerebellum Development in Newborn Rats. Neonatology 2024; 122:20-26. [PMID: 39053436 DOI: 10.1159/000540077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024]
Abstract
INTRODUCTION Preterm newborns struggle with maintaining an adequate respiratory pattern; early caffeine administration is suggested to stimulate respiration and reduce bronchopulmonary dysplasia, however, its consequences on the immature cerebellum remains unknown. This study aimed to assess the impact of early caffeine administration, at standard and high doses, accompanied by supplemental oxygen on cerebellar development in an experimental model. METHODS Five groups of Wistar pups were formed (n = 8 offspring/group): (a) negative control: no intervention; (b) placebo: pups remaining from birth until the 7th day of life (DOL) exposed to fractional inspired oxygen (FiO2) 45%, resembling preterm infant condition and as a placebo, 0.2 mL oral 5% dextrose, from the first DOL until the 14th DOL; (c) caffeine group: oral caffeine, 1st DOL 20 mg/kg, and from 2nd to 14th DOL, 5 mg/kg (standard dose); (d) caffeine at the standard dose, plus O2: during the first 7 DOLs (FiO2: 45%); (e) caffeine: 40 mg/kg in the first DOL, 10 mg/kg the next 14 DOLs, plus O2 in the first 7 DOLs (FiO2: 45%). Subjects were sacrificed on their 15th DOL; measurements were taken from the cerebellum, specifically the external granular layer (EGL) and molecular layer (ML), with quantification of cell migration. RESULTS Caffeine administration in pups resulted in a delay in cerebellum development based on persistent transitional EGL cells; this finding was exacerbated in groups exposed to caffeine plus O2, as evident from the thicker EGL. The negative control group showed near-complete cell migration with a thicker ML and a significantly smaller EGL. CONCLUSIONS Early caffeine administration in newborn rats disrupts cerebellar cortex cell processes and connectivity pathways, with exacerbated effects in groups receiving caffeine plus O2.
Collapse
Affiliation(s)
- Lourdes Lemus-Varela
- Neonatal Intensive Care Unit, Pediatric Hospital, Western Medical Center, Mexican Social Security Institute, Guadalajara, Mexico
- Ibero-American Society of Neonatology (SIBEN), Wellington, Florida, USA
| | - Blanca Torres-Mendoza
- Neurosciences Division, Western Biomedical Research Center, Institute of Mexican Social Security, Guadalajara, Mexico
- Department of Philosophical and Methodological Disciplines, Health Science Center, University of Guadalajara, Guadalajara, Mexico
| | - Paola Rabago-Domingo
- Neonatal Intensive Care Unit, Pediatric Hospital, Western Medical Center, Mexican Social Security Institute, Guadalajara, Mexico
| | - Jhonathan Cárdenas-Bedoya
- Neurosciences Division, Western Biomedical Research Center, Institute of Mexican Social Security, Guadalajara, Mexico
- Department of Philosophical and Methodological Disciplines, Health Science Center, University of Guadalajara, Guadalajara, Mexico
| | - Guillermo M Zúñiga-González
- Molecular Medicine Division, Western Biomedical Research Center, Institute of Mexican Social Security, Guadalajara, Mexico
| | - Erandis D Torres-Sanchez
- Department of Medical and Life Sciences, "La Cienega",Center, University of Guadalajara, Ocotlan, Mexico
| | - Genaro Gabriel-Ortiz
- Department of Philosophical and Methodological Disciplines, Health Science Center, University of Guadalajara, Guadalajara, Mexico
| |
Collapse
|
10
|
Hossain MK, Kim HR, Chae HJ. Aging phenotype in AD brain organoids: Track to success and challenges. Ageing Res Rev 2024; 96:102256. [PMID: 38460555 DOI: 10.1016/j.arr.2024.102256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 02/19/2024] [Accepted: 02/27/2024] [Indexed: 03/11/2024]
Abstract
Alzheimer's disease (AD) poses a complex challenge, with abnormal protein accumulation in the brain causing memory loss and cognitive decline. Traditional models fall short in AD research, prompting interest in 3D brain organoids (BOs) from human stem cells. These findings hold promise for unveiling the mechanisms of AD, especially in relation to aging. However, an understanding of the aging impact of AD remains elusive. BOs offer insight but face challenges. This review delves into the role of BOs in deciphering aging-related AD and acknowledges limitations. Strategies to enhance BOs for accurate aging modeling in AD brains are suggested. Strengthened by molecular advancements, BOs have the potential to uncover the aging phenotype, advancing AD research.
Collapse
Affiliation(s)
| | - Hyung-Ryong Kim
- Department of Pharmacology, College of Dentistry, Jeonbuk National University, Jeonju 54896, Republic of Korea.
| | - Han Jung Chae
- School of Pharmacy, Jeonbuk National University, Jeonju 54896, Republic of Korea.
| |
Collapse
|
11
|
吴 晨, 王 孟, 周 荣. [Recent research on the role of oxidative stress in the pathogenesis of attention deficit hyperactivity disorder]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:201-206. [PMID: 38436320 PMCID: PMC10921868 DOI: 10.7499/j.issn.1008-8830.2309039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/29/2023] [Indexed: 03/05/2024]
Abstract
Attention deficit hyperactivity disorder (ADHD) is a common neurodevelopmental disorder in children and adolescents, and its etiology and pathogenesis are still unclear. Brain is the organ with the largest oxygen consumption in human body and is easily affected by oxidative imbalance. Oxidative stress has become the key research direction for the pathogenesis of ADHD, but there is still a lack of relevant studies in China. Based on the latest research findings in China and overseas, this article reviews the clinical and experimental studies on oxidative stress in ADHD and explores the association of oxidative stress with neurotransmitter imbalance, neuroinflammation, and cell apoptosis in the pathogenesis of ADHD, so as to provide new research ideas for exploring the pathogenesis of ADHD.
Collapse
|
12
|
Maiese K. Mitochondria, Mitophagy, Mitoptosis, and Programmed Cell Death: Implications from Aging to Cancer. Curr Neurovasc Res 2024; 21:1-5. [PMID: 38251666 DOI: 10.2174/1567202621999240118155618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
|
13
|
Maiese K. Cornerstone Cellular Pathways for Metabolic Disorders and Diabetes Mellitus: Non-Coding RNAs, Wnt Signaling, and AMPK. Cells 2023; 12:2595. [PMID: 37998330 PMCID: PMC10670256 DOI: 10.3390/cells12222595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 11/25/2023] Open
Abstract
Metabolic disorders and diabetes (DM) impact more than five hundred million individuals throughout the world and are insidious in onset, chronic in nature, and yield significant disability and death. Current therapies that address nutritional status, weight management, and pharmacological options may delay disability but cannot alter disease course or functional organ loss, such as dementia and degeneration of systemic bodily functions. Underlying these challenges are the onset of aging disorders associated with increased lifespan, telomere dysfunction, and oxidative stress generation that lead to multi-system dysfunction. These significant hurdles point to the urgent need to address underlying disease mechanisms with innovative applications. New treatment strategies involve non-coding RNA pathways with microRNAs (miRNAs) and circular ribonucleic acids (circRNAs), Wnt signaling, and Wnt1 inducible signaling pathway protein 1 (WISP1) that are dependent upon programmed cell death pathways, cellular metabolic pathways with AMP-activated protein kinase (AMPK) and nicotinamide, and growth factor applications. Non-coding RNAs, Wnt signaling, and AMPK are cornerstone mechanisms for overseeing complex metabolic pathways that offer innovative treatment avenues for metabolic disease and DM but will necessitate continued appreciation of the ability of each of these cellular mechanisms to independently and in unison influence clinical outcome.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, NY 10022, USA
| |
Collapse
|
14
|
Maiese K. The impact of aging and oxidative stress in metabolic and nervous system disorders: programmed cell death and molecular signal transduction crosstalk. Front Immunol 2023; 14:1273570. [PMID: 38022638 PMCID: PMC10663950 DOI: 10.3389/fimmu.2023.1273570] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Life expectancy is increasing throughout the world and coincides with a rise in non-communicable diseases (NCDs), especially for metabolic disease that includes diabetes mellitus (DM) and neurodegenerative disorders. The debilitating effects of metabolic disorders influence the entire body and significantly affect the nervous system impacting greater than one billion people with disability in the peripheral nervous system as well as with cognitive loss, now the seventh leading cause of death worldwide. Metabolic disorders, such as DM, and neurologic disease remain a significant challenge for the treatment and care of individuals since present therapies may limit symptoms but do not halt overall disease progression. These clinical challenges to address the interplay between metabolic and neurodegenerative disorders warrant innovative strategies that can focus upon the underlying mechanisms of aging-related disorders, oxidative stress, cell senescence, and cell death. Programmed cell death pathways that involve autophagy, apoptosis, ferroptosis, and pyroptosis can play a critical role in metabolic and neurodegenerative disorders and oversee processes that include insulin resistance, β-cell function, mitochondrial integrity, reactive oxygen species release, and inflammatory cell activation. The silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), AMP activated protein kinase (AMPK), and Wnt1 inducible signaling pathway protein 1 (WISP1) are novel targets that can oversee programmed cell death pathways tied to β-nicotinamide adenine dinucleotide (NAD+), nicotinamide, apolipoprotein E (APOE), severe acute respiratory syndrome (SARS-CoV-2) exposure with coronavirus disease 2019 (COVID-19), and trophic factors, such as erythropoietin (EPO). The pathways of programmed cell death, SIRT1, AMPK, and WISP1 offer exciting prospects for maintaining metabolic homeostasis and nervous system function that can be compromised during aging-related disorders and lead to cognitive impairment, but these pathways have dual roles in determining the ultimate fate of cells and organ systems that warrant thoughtful insight into complex autofeedback mechanisms.
Collapse
Affiliation(s)
- Kenneth Maiese
- Innovation and Commercialization, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
15
|
Maiese K. Cognitive Impairment in Multiple Sclerosis. Bioengineering (Basel) 2023; 10:871. [PMID: 37508898 PMCID: PMC10376413 DOI: 10.3390/bioengineering10070871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Almost three million individuals suffer from multiple sclerosis (MS) throughout the world, a demyelinating disease in the nervous system with increased prevalence over the last five decades, and is now being recognized as one significant etiology of cognitive loss and dementia. Presently, disease modifying therapies can limit the rate of relapse and potentially reduce brain volume loss in patients with MS, but unfortunately cannot prevent disease progression or the onset of cognitive disability. Innovative strategies are therefore required to address areas of inflammation, immune cell activation, and cell survival that involve novel pathways of programmed cell death, mammalian forkhead transcription factors (FoxOs), the mechanistic target of rapamycin (mTOR), AMP activated protein kinase (AMPK), the silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), and associated pathways with the apolipoprotein E (APOE-ε4) gene and severe acute respiratory syndrome coronavirus (SARS-CoV-2). These pathways are intertwined at multiple levels and can involve metabolic oversight with cellular metabolism dependent upon nicotinamide adenine dinucleotide (NAD+). Insight into the mechanisms of these pathways can provide new avenues of discovery for the therapeutic treatment of dementia and loss in cognition that occurs during MS.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, NY 10022, USA
| |
Collapse
|
16
|
Liu X, Huang R, Wan J. Puerarin: a potential natural neuroprotective agent for neurological disorders. Biomed Pharmacother 2023; 162:114581. [PMID: 36966665 DOI: 10.1016/j.biopha.2023.114581] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/18/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Puerarin is an isoflavone compound derived from Pueraria lobata in traditional Chinese medicine. Accumulating evidence has indicated that puerarin demonstrates multiple pharmacological effects and exhibits treatment potential for various neurological disorders. Based on the latest research progress on puerarin as a neuroprotective agent, its pharmacological activity, molecular mechanism, and therapeutic application were systematically reviewed with emphasis on pre-clinical studies. The related information was extracted and compiled from major scientific databases, including PubMed, ScienceDirect, SpringerLink, and Chinese National Knowledge Infrastructure, using 'Puerarin', 'Neuroprotection', 'Apoptosis', 'Autophagy', 'Antioxidant', 'Mitochondria', 'Anti-inflammation' as keywords. This review complied with The Preferred Reporting Items for Systematic Reviews criteria. Forty-three articles met established inclusion and exclusion criteria. Puerarin has shown neuroprotective effects against a variety of neurological disorders, including ischemic cerebrovascular disease, subarachnoid hemorrhage, epilepsy, cognitive disorders, traumatic brain injury, Parkinson's disease, Alzheimer's disease, anxiety, depression, diabetic neuropathy, and neuroblastoma/glioblastoma. Puerarin demonstrates anti-apoptosis, proinflammatory mediator inhibitory, autophagy regulatory, anti-oxidative stress, mitochondria protection, Ca2+ influx inhibitory, and anti-neurodegenerative activities. Puerarin exerts noticeable neuroprotective effects on various models of neurological disorders in vivo (animal). This review will contribute to the development of puerarin as a novel clinical drug candidate for the treatment of neurological disorders. However, well-designed, high-quality, large-scale, multicenter randomized clinical studies are needed to determine the safety and clinical utility of puerarin in patients with neurological disorders.
Collapse
Affiliation(s)
- Xue Liu
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Rui Huang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jiye Wan
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
17
|
Oyefeso FA, Goldberg G, Opoku NYPS, Vazquez M, Bertucci A, Chen Z, Wang C, Muotri AR, Pecaut MJ. Effects of acute low-moderate dose ionizing radiation to human brain organoids. PLoS One 2023; 18:e0282958. [PMID: 37256873 PMCID: PMC10231836 DOI: 10.1371/journal.pone.0282958] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 02/27/2023] [Indexed: 06/02/2023] Open
Abstract
Human exposure to low-to-moderate dose ionizing radiation (LMD-IR) is increasing via environmental, medical, occupational sources. Acute exposure to LMD-IR can cause subclinical damage to cells, resulting in altered gene expression and cellular function within the human brain. It has been difficult to identify diagnostic and predictive biomarkers of exposure using traditional research models due to factors including lack of 3D structure in monolayer cell cultures, limited ability of animal models to accurately predict human responses, and technical limitations of studying functional human brain tissue. To address this gap, we generated brain/cerebral organoids from human induced pluripotent stem cells to study the radiosensitivity of human brain cells, including neurons, astrocytes, and oligodendrocytes. While organoids have become popular models for studying brain physiology and pathology, there is little evidence to confirm that exposing brain organoids to LMD-IR will recapitulate previous in vitro and in vivo observations. We hypothesized that exposing brain organoids to proton radiation would (1) cause a time- and dose-dependent increase in DNA damage, (2) induce cell type-specific differences in radiosensitivity, and (3) increase expression of oxidative stress and DNA damage response genes. Organoids were exposed to 0.5 or 2 Gy of 250 MeV protons and samples were collected at 30 minute, 24 hour, and 48 hour timepoints. Using immunofluorescence and RNA sequencing, we found time- and dose-dependent increases in DNA damage in irradiated organoids; no changes in cell populations for neurons, oligodendrocytes, and astrocytes by 24 hours; decreased expression of genes related to oligodendrocyte lineage, astrocyte lineage, mitochondrial function, and cell cycle progression by 48 hours; increased expression of genes related to neuron lineage, oxidative stress, and DNA damage checkpoint regulation by 48 hours. Our findings demonstrate the possibility of using organoids to characterize cell-specific radiosensitivity and early radiation-induced gene expression changes within the human brain, providing new avenues for further study of the mechanisms underlying acute neural cell responses to IR exposure at low-to-moderate doses.
Collapse
Affiliation(s)
- Foluwasomi A. Oyefeso
- Department of Biomedical Engineering Sciences, School of Medicine, Loma Linda University, Loma Linda, California, United States of America
| | - Gabriela Goldberg
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Nana Yaa P. S. Opoku
- Department of Biomedical Engineering Sciences, School of Medicine, Loma Linda University, Loma Linda, California, United States of America
| | - Marcelo Vazquez
- Departments of Pediatrics and Cellular & Molecular Medicine, School of Medicine, Center for Academic Research and Training in Anthropogeny (CARTA), Kavli Institute for Brain and Mind, Archealization Center (ArchC), University of California San Diego, La Jolla, California, United States of America
- Center for Genomics, School of Medicine, Loma Linda University, Loma Linda, California, United States of America
| | - Antonella Bertucci
- Center for Genomics, School of Medicine, Loma Linda University, Loma Linda, California, United States of America
| | - Zhong Chen
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California, United States of America
| | - Charles Wang
- Departments of Pediatrics and Cellular & Molecular Medicine, School of Medicine, Center for Academic Research and Training in Anthropogeny (CARTA), Kavli Institute for Brain and Mind, Archealization Center (ArchC), University of California San Diego, La Jolla, California, United States of America
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California, United States of America
| | - Alysson R. Muotri
- Department of Radiation Medicine, School of Medicine, Loma Linda University, Loma Linda, California, United States of America
| | - Michael J. Pecaut
- Department of Biomedical Engineering Sciences, School of Medicine, Loma Linda University, Loma Linda, California, United States of America
- Departments of Pediatrics and Cellular & Molecular Medicine, School of Medicine, Center for Academic Research and Training in Anthropogeny (CARTA), Kavli Institute for Brain and Mind, Archealization Center (ArchC), University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
18
|
El-Sayed NS, Elatrebi S, Said R, Ibrahim HF, Omar EM. Potential mechanisms underlying the association between type II diabetes mellitus and cognitive dysfunction in rats: a link between miRNA-21 and Resveratrol's neuroprotective action. Metab Brain Dis 2022; 37:2375-2388. [PMID: 35781592 PMCID: PMC9581846 DOI: 10.1007/s11011-022-01035-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 06/13/2022] [Indexed: 11/10/2022]
Abstract
Cognitive impairment is considered as a typical feature of neurodegenerative diseases in diabetes mellitus (DM). However, the exact link between cognitive dysfunction and diabetes mellitus is still vague. This study aims to investigate some of the mechanisms underlying cognitive impairment that associates diabetes mellitus and insulin resistance. We investigated the role of resveratrol as well on cognitive function in experimentally induced type 2 diabetes highlighting on its influence on the expression of brain miRNA 21. Resveratrol is a naturally occurring, biologically active compound that has numerous significant impacts on the body. Type 2 diabetes mellitus was induced by high fat diet followed a single dose of streptozotocin. Diabetic rats were treated with resveratrol for four weeks. Rats were sacrificed after neurobehavioral testing. Hippocampal tissues were used to assess expression of miRNA 21, GSK and oxidative stress markers. Serum samples were obtained to determine glucose levels, lipid profile and insulin levels. Hippocampal and serum AGEs were measured as well and HOMA IR was calculated. We detected memory impairment and disturbed insulin signaling in diabetic rats. These derangements were reversed by resveratrol treatment partially due to increased expression of miRNA-21. Our study pins the role of miRNA-21 in modulating brain insulin signaling and hence alleviating cognitive dysfunction accompanying diabetes mellitus.
Collapse
Affiliation(s)
- Norhan S. El-Sayed
- Department of Medical Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Soha Elatrebi
- Department of Clinical Pharmacology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Rasha Said
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Heba F. Ibrahim
- Department of Histology and Cell Biology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Eman M. Omar
- Department of Medical Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
19
|
A Mouse Model of Neurodegeneration Induced by Blade Penetrating Stab Wound to the Hippocampus. BIOLOGY 2022; 11:biology11091365. [PMID: 36138848 PMCID: PMC9495452 DOI: 10.3390/biology11091365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 08/28/2022] [Accepted: 09/14/2022] [Indexed: 12/02/2022]
Abstract
Simple Summary To date, various animal models of traumatic brain injury (TBI) have been developed to investigate the cellular and molecular networks underlying the pathogenesis of neurodegeneration, and to examine the safeness and therapeutic efficacy of drugs. However, the commonly used animal TBI models usually have the weaknesses of low reproducibility and high mortality rates. In the present study, we created a mouse model of cognitive deficits induced by a blade penetrating stab wound to the hippocampus (HBSI). This model will contribute a better understanding of neurodegeneration and accelerate the translation of preclinical research to clinical applications. Abstract Traumatic brain injury (TBI) is closely associated with the later development of neurodegenerative and psychiatric diseases which are still incurable. Although various animal TBI models have been generated, they usually have weaknesses in standardization, survivability and/or reproducibility. In the present study, we investigated whether applying a blade penetrating stab wound to the hippocampus would create an animal model of cognitive deficits. Open-field, Morris water maze and Barnes maze tests were used to evaluate the animal behaviors. The immunofluorescence staining of NeuN, GFAP, IBA1, and TUNEL was conducted to analyze the changes in neurons, astrocytes, and microglia, as well as cell death. Mice with a hippocampal blade stab injury (HBSI) displayed the activation of microglia and astrocytes, inflammation, neuronal apoptosis, and deficits in spatial learning and memory. These findings suggest that HBSI is an easy approach to generate a reliable in vivo model of TBI to capture hemorrhage, neuroinflammation, reactive gliosis, and neural death, as well as cognitive deficits observed in human patients.
Collapse
|
20
|
Dim Blue Light at Night Induces Spatial Memory Impairment in Mice by Hippocampal Neuroinflammation and Oxidative Stress. Antioxidants (Basel) 2022; 11:antiox11071218. [PMID: 35883709 PMCID: PMC9311634 DOI: 10.3390/antiox11071218] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/12/2022] [Accepted: 06/17/2022] [Indexed: 12/04/2022] Open
Abstract
Light pollution is one of the most serious public problems, especially the night light. However, the effect of dim blue light at night (dLAN-BL) on cognitive function is unclear. In this study, we evaluated the effects of exposure to dLAN-BL in C57BL/6J mice for 4 consecutive weeks. Our results showed dLAN-BL significantly impaired spatial learning and memory and increased plasma corticosterone level in mice. Consistent with these changes, we observed dLAN-BL significantly increased the numbers and activation of microglia and the levels of oxidative stress product MDA in the hippocampus, decreased the levels of antioxidant enzymes Glutathione peroxidase (GSH-Px), Superoxide dismutase (SOD), Gluathione reductase (Gsr), total antioxidants (T-AOC) and the number of neurons in the hippocampus, up-regulated the mRNA expression levels of IL6, TNF-α and the protein expression levels of iNOS, COX2, TLR4, p-p65, Cleaved-Caspase3 and BAX, and down-regulated the mRNA expression levels of IL4, IL10, Psd95, Snap25, Sirt1, Dcx and the protein expression level of BCL2. In vitro results further showed corticosterone (10uM)-induced BV2 cell activation and up-regulated content of IL6, TNF-α in the cell supernatant and the protein expression levels of iNOS, COX2, p-p65 in BV2 cells. Our findings suggested dLAN-BL up-regulated plasma corticosterone level and hippocampal microglia activation, which in turn caused oxidative stress and neuroinflammation, leading to neuronal loss and synaptic dysfunction, ultimately leading to spatial learning and memory dysfunction in mice.
Collapse
|