1
|
Gao MZ, Zeng JY, Chen XJ, Shi L, Hong FY, Lin M, Luo JW, Chen H. Dimethyl fumarate ameliorates oxidative stress-induced acute kidney injury after traumatic brain injury by activating Keap1-Nrf2/HO-1 signaling pathway. Heliyon 2024; 10:e32377. [PMID: 38947486 PMCID: PMC11214498 DOI: 10.1016/j.heliyon.2024.e32377] [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: 02/14/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 07/02/2024] Open
Abstract
Acute kidney injury (AKI) frequently emerges as a consequential non-neurological sequel to traumatic brain injury (TBI), significantly contributing to heightened mortality risks. The intricate interplay of oxidative stress in the pathophysiology of TBI underscores the centrality of the Keap1-Nrf2/HO-1 signaling pathway as a pivotal regulator in this context. This study endeavors to elucidate the involvement of the Keap1-Nrf2/HO-1 pathway in modulating oxidative stress in AKI subsequent to TBI and concurrently explore the therapeutic efficacy of dimethyl fumarate (DMF). A rat model of TBI was established via the Feeney free-fall method, incorporating interventions with varying concentrations of DMF. Assessment of renal function ensued through measurements of serum creatinine and neutrophil gelatinase-associated lipocalin. Morphological evaluation of renal pathology was conducted employing quantitative hematoxylin and eosin staining. The inflammatory response was scrutinized by quantifying interleukin (IL)-6, IL-1β, and tumor necrosis factor-α levels. Oxidative stress levels were discerned through quantification of malondialdehyde and superoxide dismutase. The apoptotic cascade was examined via the terminal deoxynucleotidyl transferase dUTP deletion labeling assay. Western blotting provided insights into the expression dynamics of proteins affiliated with the Keap1-Nrf2/HO-1 pathway and apoptosis. The findings revealed severe kidney injury, heightened oxidative stress, inflammation, and apoptosis in the traumatic brain injury model. Treatment with DMF effectively reversed these changes, alleviating oxidative stress by activating the Keap1-Nrf2/HO-1 signaling pathway, ultimately conferring protection against AKI. Activating Keap1-Nrf2/HO-1 signaling pathway may be a potential therapeutic strategy for attenuating oxidative stress-induced AKI after TBI.
Collapse
Affiliation(s)
- Mei-zhu Gao
- Department of Nephrology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Jing-yi Zeng
- Department of Nephrology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Xue-jing Chen
- Department of Nephrology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Lan Shi
- Department of Intensive Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Fu-yuan Hong
- Department of Nephrology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Miao Lin
- Department of Nephrology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Jie-wei Luo
- Department of Traditional Chinese Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Han Chen
- The Fourth Department of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Provincial Key Laboratory of Critical Care Medicine, Fuzhou, Fujian, 350001, China
| |
Collapse
|
2
|
Fesharaki-Zadeh A. Oxidative Stress in Traumatic Brain Injury. Int J Mol Sci 2022; 23:ijms232113000. [PMID: 36361792 PMCID: PMC9657447 DOI: 10.3390/ijms232113000] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 11/17/2022] Open
Abstract
Traumatic Brain Injury (TBI) remains a major cause of disability worldwide. It involves a complex neurometabolic cascade, including oxidative stress. The products of this manuscript is examining the underlying pathophysiological mechanism, including reactive oxygen species (ROS) and reactive nitrogen species (RNS). This process in turn leads to secondary injury cascade, which includes lipid peroxidation products. These reactions ultimately play a key role in chronic inflammation and synaptic dysfunction in a synergistic fashion. Although there are no FDA approved antioxidant therapy for TBI, there is a number of antioxidant therapies that have been tested and include free radical scavengers, activators of antioxidant systems, inhibitors of free radical generating enzymes, and antioxidant enzymes. Antioxidant therapies have led to cognitive and functional recovery post TBI, and they offer a promising treatment option for patients recovering from TBI. Current major challenges in treatment of TBI symptoms include heterogenous nature of injury, as well as access to timely treatment post injury. The inherent benefits of antioxidant therapies include minimally reported side effects, and relative ease of use in the clinical setting. The current review also provides a highlight of the more studied anti-oxidant regimen with applicability for TBI treatment with potential use in the real clinical setting.
Collapse
Affiliation(s)
- Arman Fesharaki-Zadeh
- Yale School of Medicine, Department of Neurology, Yale University, New Haven, CT 06510, USA
| |
Collapse
|
3
|
Reprint of: Oxygen Free Radicals and Iron in Relation to Biology and Medicine: Some Problems and Concepts. Arch Biochem Biophys 2022; 726:109246. [PMID: 35680438 DOI: 10.1016/j.abb.2022.109246] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
4
|
Binvignat O, Olloquequi J. Excitotoxicity as a Target Against Neurodegenerative Processes. Curr Pharm Des 2020; 26:1251-1262. [PMID: 31931694 DOI: 10.2174/1381612826666200113162641] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/27/2019] [Indexed: 12/20/2022]
Abstract
The global burden of neurodegenerative diseases is alarmingly increasing in parallel to the aging of population. Although the molecular mechanisms leading to neurodegeneration are not completely understood, excitotoxicity, defined as the injury and death of neurons due to excessive or prolonged exposure to excitatory amino acids, has been shown to play a pivotal role. The increased release and/or decreased uptake of glutamate results in dysregulation of neuronal calcium homeostasis, leading to oxidative stress, mitochondrial dysfunctions, disturbances in protein turn-over and neuroinflammation. Despite the anti-excitotoxic drug memantine has shown modest beneficial effects in some patients with dementia, to date, there is no effective treatment capable of halting or curing neurodegenerative diseases such as Alzheimer's disease, Parkinson disease, Huntington's disease or amyotrophic lateral sclerosis. This has led to a growing body of research focusing on understanding the mechanisms associated with the excitotoxic insult and on uncovering potential therapeutic strategies targeting these mechanisms. In the present review, we examine the molecular mechanisms related to excitotoxic cell death. Moreover, we provide a comprehensive and updated state of the art of preclinical and clinical investigations targeting excitotoxic- related mechanisms in order to provide an effective treatment against neurodegeneration.
Collapse
Affiliation(s)
| | - Jordi Olloquequi
- Laboratory of Cellular and Molecular Pathology, Instituto de Ciencias Biomedicas, Facultad de Ciencias de la Salud, Universidad Autonoma de Chile, Talca, Chile
| |
Collapse
|
5
|
Efe A, Neşelioğlu S, Soykan A. An Investigation of the Dynamic Thiol/Disulfide Homeostasis, As a Novel Oxidative Stress Plasma Biomarker, in Children With Autism Spectrum Disorders. Autism Res 2020; 14:473-487. [PMID: 33210838 DOI: 10.1002/aur.2436] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 10/30/2020] [Accepted: 11/03/2020] [Indexed: 12/26/2022]
Abstract
We aimed to investigate the role of impaired oxidant-antioxidant homeostasis on the etiopathogenesis of autism with a novel oxidative stress (OS) marker, dynamic thiol/disulfide homeostasis (DTDH), and relationship between the symptom severity and markers. A total of 60 children with ASD aged 3-10 years and 54 unaffected children were investigated for the plasma DTDH parameters. A sociodemographic-data form, K-SADS-PL, Childhood Autism Rating Scale, Abnormal Behavior Checklist, Autism Behavior Checklist, and a developmentally appropriate IQ test were administered to all participants. Distortion of DTDH to the OS-side in the autism group was determined with lower plasma levels of native and total thiol, in contrast to a higher disulfide and thiol oxidation-reduction ratio. However, biomarkers had no correlation with the symptom severity of autism. Cutoff values for each parameter on the ROC curve might be useful to predict ASD and each DTDH biomarker was detected as an independent predictor of ASD. The present study demonstrated a disturbed redox status and absence of an expected compensatory increase in antioxidant response in a pediatric sample of ASD by measuring dynamic oxidation/reduction shifts with a novel, practical and reproducible analytical technique, and contributes to data regarding oxidative hypothesis on autism and raises the question of the place of antioxidants in autism treatment. Our results may suggest predictive usefulness of the plasma DTDH biomarkers in ASD, despite the study being conducted with a modestly small sample size that makes further research with a larger replication sample necessary to substantiate the findings. LAY SUMMARY: Dynamic thiol/disulfide homeostasis is a novel plasma marker used to determine the oxidative stress which is a natural result of disequilibrium between the oxidants and antioxidants in the human body. There is increasing interest regarding a central biological linking role of oxidative stress among the other etiological factors of autism. Our findings on the disturbed plasma dynamic thiol/disulfide homeostasis in children with autism and the absence of an expected antioxidant response against increased oxidative stress supports the data concerning the role of oxidative stress on the etiology of autism and the need of further research on the place of antioxidants in autism treatment.
Collapse
Affiliation(s)
- Ayşegül Efe
- Department of Child and Adolescent Psychiatry, Dr. Sami Ulus Gynecology Obstetrics and Child Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Salim Neşelioğlu
- Department of Medical Biochemistry, Yıldırım Beyazıt University, Ankara Atatürk Training and Research Hospital, Ankara, Turkey
| | - Ayla Soykan
- Department of Child and Adolescent Psychiatry, Ankara University, School of Medicine, Ankara, Turkey
| |
Collapse
|
6
|
Revisiting Traumatic Brain Injury: From Molecular Mechanisms to Therapeutic Interventions. Biomedicines 2020; 8:biomedicines8100389. [PMID: 33003373 PMCID: PMC7601301 DOI: 10.3390/biomedicines8100389] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 09/25/2020] [Accepted: 09/26/2020] [Indexed: 12/15/2022] Open
Abstract
Studying the complex molecular mechanisms involved in traumatic brain injury (TBI) is crucial for developing new therapies for TBI. Current treatments for TBI are primarily focused on patient stabilization and symptom mitigation. However, the field lacks defined therapies to prevent cell death, oxidative stress, and inflammatory cascades which lead to chronic pathology. Little can be done to treat the mechanical damage that occurs during the primary insult of a TBI; however, secondary injury mechanisms, such as inflammation, blood-brain barrier (BBB) breakdown, edema formation, excitotoxicity, oxidative stress, and cell death, can be targeted by therapeutic interventions. Elucidating the many mechanisms underlying secondary injury and studying targets of neuroprotective therapeutic agents is critical for developing new treatments. Therefore, we present a review on the molecular events following TBI from inflammation to programmed cell death and discuss current research and the latest therapeutic strategies to help understand TBI-mediated secondary injury.
Collapse
|
7
|
The Role of Oxidative Stress and Bioenergetic Dysfunction in Sulfite Oxidase Deficiency: Insights from Animal Models. Neurotox Res 2018; 35:484-494. [DOI: 10.1007/s12640-018-9986-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/13/2018] [Accepted: 11/26/2018] [Indexed: 02/07/2023]
|
8
|
Jhelum P, Karisetty BC, Kumar A, Chakravarty S. Implications of Epigenetic Mechanisms and their Targets in Cerebral Ischemia Models. Curr Neuropharmacol 2018; 15:815-830. [PMID: 27964703 PMCID: PMC5652028 DOI: 10.2174/1570159x14666161213143907] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 11/07/2016] [Accepted: 12/09/2016] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Understanding the complexities associated with the ischemic condition and identifying therapeutic targets in ischemia is a continued challenge in stroke biology. Emerging evidence reveals the potential involvement of epigenetic mechanisms in the incident and outcome of stroke, suggesting novel therapeutic options of targeting different molecules related to epigenetic regulation. OBJECTIVE This review summarizes our current understanding of ischemic pathophysiology, describes various in vivo and in vitro models of ischemia, and examines epigenetic modifications associated with the ischemic condition. METHOD We focus on microRNAs, DNA methylation, and histone modifying enzymes, and present how epigenetic studies are revealing novel drug target candidates in stroke. CONCLUSION Finally, we discuss emerging approaches for the prevention and treatment of stroke and post-stroke effects using pharmacological interventions with a wide therapeutic window.
Collapse
Affiliation(s)
- Priya Jhelum
- Chemical Biology, CSIR, Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad 500007, India
| | - Bhanu C Karisetty
- Chemical Biology, CSIR, Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad 500007, India
| | - Arvind Kumar
- CSIR, Centre for Cellular and Molecular Biology, Habsiguda, Uppal Road, Hyderabad 500007, India
| | - Sumana Chakravarty
- Chemical Biology, CSIR-Indian Institute of Chemical Technology (IICT), Tarnaka, Hyderabad-500007, India
| |
Collapse
|
9
|
Effects of Female Sex Steroids Administration on Pathophysiologic Mechanisms in Traumatic Brain Injury. Transl Stroke Res 2017; 9:393-416. [PMID: 29151229 DOI: 10.1007/s12975-017-0588-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/16/2017] [Accepted: 11/07/2017] [Indexed: 12/19/2022]
Abstract
Secondary brain damage following initial brain damage in traumatic brain injury (TBI) is a major cause of adverse outcomes. There are many gaps in TBI research and a lack of therapy to limit debilitating outcomes in TBI or enhance the neurogenesis, despite pre-clinical and clinical research performed in TBI. Females show harmful outcomes against brain damage including TBI less than males, independent of different TBI occurrence. A significant reduction in secondary brain damage and improvement in neurologic outcome post-TBI has been reported following the use of progesterone and estrogen in many experimental studies. Although useful features of sex steroids including progesterone have been identified in TBI clinical trials I and II, clinical trials III have been unsuccessful. This review article focuses on evidence of secondary injury mechanisms and neuroprotective effects of estrogen and progesterone in TBI. Understanding these mechanisms may enable researchers to achieve greater success in TBI clinical studies. It seems that the design of clinical studies should be revised due to translation loss of animal studies to clinical studies. The heterogeneous and complex nature of TBI, the endogenous levels of sex hormones at the time of taking these hormones, the therapeutic window of the drug, the dosage of the drug, the selection of appropriate targets in evaluation, the determination of responsive population, gender and age based on animal studies should be considered in the design of TBI human studies in future.
Collapse
|
10
|
Individual Amino Acid Supplementation Can Improve Energy Metabolism and Decrease ROS Production in Neuronal Cells Overexpressing Alpha-Synuclein. Neuromolecular Med 2017. [PMID: 28620826 DOI: 10.1007/s12017-017-8448-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by alpha-synuclein accumulation and loss of dopaminergic neurons in the substantia nigra (SN) region of the brain. Increased levels of alpha-synuclein have been shown to result in loss of mitochondrial electron transport chain complex I activity leading to increased reactive oxygen species (ROS) production. WT alpha-synuclein was stably overexpressed in human BE(2)-M17 neuroblastoma cells resulting in increased levels of an alpha-synuclein multimer, but no increase in alpha-synuclein monomer levels. Oxygen consumption was decreased by alpha-synuclein overexpression, but ATP levels did not decrease and ROS levels did not increase. Treatment with ferrous sulfate, a ROS generator, resulted in decreased oxygen consumption in both control and alpha-synuclein overexpressing cells. However, this treatment only decreased ATP levels and increased ROS production in the cells overexpressing alpha-synuclein. Similarly, paraquat, another ROS generator, decreased ATP levels in the alpha-synuclein overexpressing cells, but not in the control cells, further demonstrating how alpha-synuclein sensitized the cells to oxidative insult. Proteomic analysis yielded molecular insights into the cellular adaptations to alpha-synuclein overexpression, such as the increased abundance of many mitochondrial proteins. Many amino acids and citric acid cycle intermediates and their ester forms were individually supplemented to the cells with L-serine, L-proline, L-aspartate, or L-glutamine decreasing ROS production in oxidatively stressed alpha-synuclein overexpressing cells, while diethyl oxaloacetate or L-valine supplementation increased ATP levels. These results suggest that dietary supplementation with individual metabolites could yield bioenergetic improvements in PD patients to delay loss of dopaminergic neurons.
Collapse
|
11
|
Dadhania VP, Trivedi PP, Vikram A, Tripathi DN. Nutraceuticals against Neurodegeneration: A Mechanistic Insight. Curr Neuropharmacol 2017; 14:627-40. [PMID: 26725888 PMCID: PMC4981739 DOI: 10.2174/1570159x14666160104142223] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 08/17/2015] [Accepted: 01/01/2016] [Indexed: 12/20/2022] Open
Abstract
The mechanisms underlying neurodegenerative disorders are complex and multifactorial; however, accumulating evidences suggest few common shared pathways. These common pathways include mitochondrial dysfunction, intracellular Ca2+ overload, oxidative stress and inflammation. Often multiple pathways co-exist, and therefore limit the benefits of therapeutic interventions. Nutraceuticals have recently gained importance owing to their multifaceted effects. These food-based approaches are believed to target multiple pathways in a slow but more physiological manner without causing severe adverse effects. Available information strongly supports the notion that apart from preventing the onset of neuronal damage, nutraceuticals can potentially attenuate the continued progression of neuronal destruction. In this article, we i) review the common pathways involved in the pathogenesis of the toxicants-induced neurotoxicity and neurodegenerative disorders with special emphasis on Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), Multiple sclerosis (MS) and Amyotrophic lateral sclerosis (ALS), and ii) summarize current research advancements on the effects of nutraceuticals against these detrimental pathways.
Collapse
Affiliation(s)
| | | | - Ajit Vikram
- Department of Internal Medicine, The University of Iowa, Iowa City, IA-52240, USA.
| | - Durga Nand Tripathi
- DNT at Center for Translational Cancer Research, Institute of Biosciences & Technology, Texas A&M University Health Science Center, Houston, TX-77030, USA.
| |
Collapse
|
12
|
Tian T, Li LL, Zhang SQ, Ni H. Long-Term Effects of Ketogenic Diet on Subsequent Seizure-Induced Brain Injury During Early Adulthood: Relationship of Seizure Thresholds to Zinc Transporter-Related Gene Expressions. Biol Trace Elem Res 2016; 174:369-376. [PMID: 27147436 DOI: 10.1007/s12011-016-0730-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 04/28/2016] [Indexed: 12/30/2022]
Abstract
The divalent cation zinc is associated with cortical plasticity. However, the mechanism of zinc in the pathophysiology of cortical injury-associated neurobehavioral damage following neonatal seizures is uncertain. We have previously shown upregulated expression of ZnT-3; MT-3 in hippocampus of neonatal rats submitted to flurothyl-induced recurrent seizures, which was restored by pretreatment with ketogenic diet (KD). In this study, utilizing a novel "twist" seizure model by coupling early-life flurothyl-induced seizures with later exposure to penicillin, we further investigated the long-term effects of KD on cortical expression of zinc homeostasis-related genes in a systemic scale. Ten Sprague-Dawley rats were assigned each averagely into the non-seizure plus normal diet (NS + ND), non-seizure plus KD (NS + KD), recurrent seizures plus normal diet (RS + ND) and recurrent seizures plus KD (RS + KD) group. Recurrent seizures were induced by volatile flurothyl during P9-P21. During P23-P53, rats in NS + KD and RS + KD groups were dieted with KD. Neurological behavioral parameters of brain damage (plane righting reflex, cliff avoidance reflex, and open field test) were observed at P43. At P63, we examined seizure threshold using penicillin, then the cerebral cortex were evaluated for real-time RT-PCR and western blot study. The RS + ND group showed worse performances in neurological reflex tests and reduced latencies to myoclonic seizures induced by penicillin compared with the control, which was concomitant with altered expressions of ZnT-7, MT-1, MT-2, and ZIP7. Specifically, there was long-term elevated expression of ZIP7 in RS + ND group compared with that in NS + ND that was restored by chronic ketogenic diet (KD) treatment in RS + KD group, which was quite in parallel with the above neurobehavioral changes. Taken together, these findings indicate that the long-term altered expression of the metal transporter ZIP7 in adult cerebral cortex might correlate with neurobehavioral damage and reduced seizure threshold following recurrent neonate seizures and further highlights ZIP7 as a candidate for therapeutic target of KD for the treatment of neonatal seizure-induced long-term brain damage.
Collapse
Affiliation(s)
- Tian Tian
- Neurology Laboratory, Institute of Pediatrics, Children' Hospital of Soochow University, No.303, Jingde Road, 215003, Suzhou, People's Republic of China
| | - Li-Li Li
- Neurology Laboratory, Institute of Pediatrics, Children' Hospital of Soochow University, No.303, Jingde Road, 215003, Suzhou, People's Republic of China
| | - Shu-Qi Zhang
- Neurology Laboratory, Institute of Pediatrics, Children' Hospital of Soochow University, No.303, Jingde Road, 215003, Suzhou, People's Republic of China
| | - Hong Ni
- Neurology Laboratory, Institute of Pediatrics, Children' Hospital of Soochow University, No.303, Jingde Road, 215003, Suzhou, People's Republic of China.
| |
Collapse
|
13
|
Sanganalmath SK, Gopal P, Parker JR, Downs RK, Parker JC, Dawn B. Global cerebral ischemia due to circulatory arrest: insights into cellular pathophysiology and diagnostic modalities. Mol Cell Biochem 2016; 426:111-127. [PMID: 27896594 DOI: 10.1007/s11010-016-2885-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 11/08/2016] [Indexed: 02/07/2023]
Abstract
Circulatory arrest (CA) remains a major unresolved public health problem in the United States; the annual incidence of which is ~0.50 to 0.55 per 1000 population. Despite seminal advances in therapeutic approaches over the past several decades, brain injury continues to be the leading cause of morbidity and mortality after CA. In brief, CA typically results in global cerebral ischemia leading to delayed neuronal death in the hippocampal pyramidal cells as well as in the cortical layers. The dynamic changes occurring in neurons after CA are still unclear, and predicting these neurological changes in the brain still remains a difficult issue. It is hypothesized that the "no-flow" period produces a cytotoxic cascade of membrane depolarization, Ca2+ ion influx, glutamate release, acidosis, and resultant activation of lipases, nucleases, and proteases. Furthermore, during reperfusion injury, neuronal death occurs due to the generation of free radicals by interfering with the mitochondrial respiratory chain. The efficacy of many pharmacological agents for CA patients has often been disappointing, reflecting our incomplete understanding of this enigmatic disease. The primary obstacles to the development of a neuroprotective therapy in CA include uncertainties with regard to the precise cause(s) of neuronal dysfunction and what to target. In this review, we summarize our knowledge of the pathophysiology as well as specific cellular changes in brain after CA and revisit the most important neurofunctional, neuroimaging techniques, and serum biomarkers as potent predictors of neurologic outcome in CA patients.
Collapse
Affiliation(s)
- Santosh K Sanganalmath
- Division of Cardiovascular Diseases, Department of Medicine, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA.
| | - Purva Gopal
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - John R Parker
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, University of Louisville, Louisville, KY, USA
| | - Richard K Downs
- Division of Neuroradiology, Department of Radiology, University of Louisville, Louisville, KY, USA
| | - Joseph C Parker
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, University of Louisville, Louisville, KY, USA
| | - Buddhadeb Dawn
- Division of Cardiovascular Diseases, Department of Medicine, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| |
Collapse
|
14
|
Zhao H, Han Z, Ji X, Luo Y. Epigenetic Regulation of Oxidative Stress in Ischemic Stroke. Aging Dis 2016; 7:295-306. [PMID: 27330844 PMCID: PMC4898926 DOI: 10.14336/ad.2015.1009] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 10/09/2015] [Indexed: 12/11/2022] Open
Abstract
The prevalence and incidence of stroke rises with life expectancy. However, except for the use of recombinant tissue-type plasminogen activator, the translation of new therapies for acute stroke from animal models into humans has been relatively unsuccessful. Oxidative DNA and protein damage following stroke is typically associated with cell death. Cause-effect relationships between reactive oxygen species and epigenetic modifications have been established in aging, cancer, acute pancreatitis, and fatty liver disease. In addition, epigenetic regulatory mechanisms during stroke recovery have been reviewed, with focuses mainly on neural apoptosis, necrosis, and neuroplasticity. However, oxidative stress-induced epigenetic regulation in vascular neural networks following stroke has not been sufficiently explored. Improved understanding of the epigenetic regulatory network upon oxidative stress may provide effective antioxidant approaches for treating stroke. In this review, we summarize the epigenetic events, including DNA methylation, histone modification, and microRNAs, that result from oxidative stress following experimental stroke in animal and cell models, and the ways in which epigenetic changes and their crosstalk influence the redox state in neurons, glia, and vascular endothelial cells, helping us to understand the foregone and vicious epigenetic regulation of oxidative stress in the vascular neural network following stroke.
Collapse
Affiliation(s)
- Haiping Zhao
- 1Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Ziping Han
- 1Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Xunming Ji
- 22Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Yumin Luo
- 1Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; 3Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
| |
Collapse
|
15
|
Krityakiarana W, Zhao PM, Nguyen K, Gomez-Pinilla F, Kotchabhakdi N, de Vellis J, Espinosa-Jeffrey A. Proof-of Concept that an Acute Trophic Factors Intervention After Spinal Cord Injury Provides an Adequate Niche for Neuroprotection, Recruitment of Nestin-Expressing Progenitors and Regeneration. Neurochem Res 2016; 41:431-49. [PMID: 26883642 PMCID: PMC5352162 DOI: 10.1007/s11064-016-1850-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 01/22/2016] [Accepted: 01/25/2016] [Indexed: 12/16/2022]
Abstract
Trophic factor treatment has been shown to improve the recovery of brain and spinal cord injury (SCI). In this study, we examined the effects of TSC1 (a combination of insulin-like growth factor 1 and transferrin) 4 and 8 h after SCI at the thoracic segment level (T12) in nestin-GFP transgenic mice. TSC1 treatment for 4 and 8 h increased the number of nestin-expressing cells around the lesion site and prevented Wallerian degeneration. Treatment with TSC1 for 4 h significantly increased heat shock protein (HSP)-32 and HSP-70 expression 1 and 2 mm from lesion site (both, caudal and rostral). Conversely, the number of HSP-32 positive cells decreased after an 8-h TSC1 treatment, although it was still higher than in both, non-treated SCI and intact spinal cord animals. Furthermore, TSC1 increased NG2 expressing cell numbers and preserved most axons intact, facilitating remyelination and repair. These results support our hypothesis that TSC1 is an effective treatment for cell and tissue neuroprotection after SCI. An early intervention is crucial to prevent secondary damage of the injured SC and, in particular, to prevent Wallerian degeneration.
Collapse
Affiliation(s)
- Warin Krityakiarana
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at University of California, Los Angeles, 635 Charles E. Young Drive South, Suite 375E, Los Angeles, CA, 90095-7332, USA.
- Division of Physiotherapy, Faculty of Health Science, Srinakharinwirot University, Bangkok, Thailand.
| | - Paul M Zhao
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at University of California, Los Angeles, 635 Charles E. Young Drive South, Suite 375E, Los Angeles, CA, 90095-7332, USA
| | - Kevin Nguyen
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at University of California, Los Angeles, 635 Charles E. Young Drive South, Suite 375E, Los Angeles, CA, 90095-7332, USA
| | - Fernando Gomez-Pinilla
- Department of Physiological Sciences and Department of Neurosurgery, University of California at Los Angeles, Los Angeles, CA, USA
- Department of Physiology, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Naiphinich Kotchabhakdi
- Neuro-Behavioural Biology Center, Institute of Science and Technology for Research and Development, Mahidol University, 999 Phutthamonthol 4 Road, Salaya, Phutthamonthol, Nakornpathom, 73170, Thailand
| | - Jean de Vellis
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at University of California, Los Angeles, 635 Charles E. Young Drive South, Suite 375E, Los Angeles, CA, 90095-7332, USA
| | - Araceli Espinosa-Jeffrey
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at University of California, Los Angeles, 635 Charles E. Young Drive South, Suite 375E, Los Angeles, CA, 90095-7332, USA.
| |
Collapse
|
16
|
Bariamis SE, Magoulas GE, Grafanaki K, Pontiki E, Tsegenidis T, Athanassopoulos CM, Maroulis G, Papaioannou D, Hadjipavlou-Litina D. Synthesis and biological evaluation of new C-10 substituted dithranol pleiotropic hybrids. Bioorg Med Chem 2015; 23:7251-63. [PMID: 26515039 DOI: 10.1016/j.bmc.2015.10.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 10/06/2015] [Accepted: 10/15/2015] [Indexed: 11/26/2022]
Abstract
Selective alkylation of the antipsoriatic drug dithranol (DTR) at C-10 with tert-butyl bromoacetate, followed by acid-mediated deprotection, produced the corresponding carboxylic acid 4 which was coupled with selectively protected polyamines (PAs), such as putrescine (PUT), spermidine (SPD) and spermine (SPM), dopamine and aliphatic amines and substituted benzylamines producing a series of DTR-PA hybrids, after acid-mediated deprotection, as well as simple amides. The compounds were tested as antioxidants and inhibitors of lipoxygenase (LOX). The amides 4,4'-dimethoxybenzhydrylamide 13 (86% and 95%), 2,4-dimethoxybenzylamide 12 (87% and 81%) and dodecylamide 9 (98% and 74%), and the hybrid DTR-SPM (7) (93% and 87%), showed the highest antioxidant activity in the DPPH and AAPH assays, whereas the most potent inhibitors of LOX were amide 13 (IC50=7 μM), the benzylamide 10 (IC50=7.9 μM) and the butylamide 8 (IC50=10 μM). Molecular binding studies showed that binding of these derivatives into the hydrophobic domain blocks approach of substrate to the active site, inhibiting soybean LOX. Amide 13 presented the highest anti-inflammatory activity (79.7%). The DTR moiety was absolutely necessary for securing high anti-inflammatory potency. Ethyl ester 3 (IC50=0.357 μM) and the amides 9 (IC50=0.022 μM) and 13 (IC50=0.56 μM) exhibited higher antiproliferative activity than DTR (IC50=0.945 μM) on HaCaT keratinocytes whereas amide 13 generally presented better cytocompatibility. Amide 13 is a very promising lead compound for further development as an anti-inflammatory and antiproliferative agent.
Collapse
Affiliation(s)
- Stavros E Bariamis
- Department of Chemistry, School of Natural Sciences, University of Patras, 26504 Patras, Greece
| | - George E Magoulas
- Department of Chemistry, School of Natural Sciences, University of Patras, 26504 Patras, Greece
| | - Katerina Grafanaki
- Laboratory of Biochemistry, Department of Medicine, School of Health Sciences, University of Patras, 26504 Patras, Greece
| | - Eleni Pontiki
- Department of Pharmaceutical Chemistry, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Theodore Tsegenidis
- Department of Chemistry, School of Natural Sciences, University of Patras, 26504 Patras, Greece
| | | | - George Maroulis
- Department of Chemistry, School of Natural Sciences, University of Patras, 26504 Patras, Greece
| | - Dionissios Papaioannou
- Department of Chemistry, School of Natural Sciences, University of Patras, 26504 Patras, Greece.
| | - Dimitra Hadjipavlou-Litina
- Department of Pharmaceutical Chemistry, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece.
| |
Collapse
|
17
|
Oxidative stress inhibition and oxidant activity by fibrous clays. Colloids Surf B Biointerfaces 2015; 133:32-5. [DOI: 10.1016/j.colsurfb.2015.05.042] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 05/20/2015] [Accepted: 05/22/2015] [Indexed: 11/18/2022]
|
18
|
Traumatic brain injury and NADPH oxidase: a deep relationship. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:370312. [PMID: 25918580 PMCID: PMC4397034 DOI: 10.1155/2015/370312] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 03/18/2015] [Indexed: 12/21/2022]
Abstract
Traumatic brain injury (TBI) represents one of the major causes of mortality and disability in the world.
TBI is characterized by primary damage resulting from the mechanical forces applied to the head as a direct result of the trauma and by the subsequent secondary injury due to a complex cascade of biochemical events that eventually lead to neuronal cell death. Oxidative stress plays a pivotal role in the genesis of the delayed harmful effects contributing to permanent damage. NADPH oxidases (Nox), ubiquitary membrane multisubunit enzymes whose unique function is the production of reactive oxygen species (ROS), have been shown to be a major source of ROS in the brain and to be involved in several neurological diseases. Emerging evidence demonstrates that Nox is upregulated after TBI, suggesting Nox critical role in the onset and development of this pathology.
In this review, we summarize the current evidence about the role of Nox enzymes in the pathophysiology of TBI.
Collapse
|
19
|
McGinn MJ, Povlishock JT. Cellular and molecular mechanisms of injury and spontaneous recovery. HANDBOOK OF CLINICAL NEUROLOGY 2015; 127:67-87. [PMID: 25702210 DOI: 10.1016/b978-0-444-52892-6.00005-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Until recently, most have assumed that traumatic brain injury (TBI) was singularly associated with the overt destruction of brain tissue resulting in subsequent morbidity or death. More recently, experimental and clinical studies have shown that the pathobiology of TBI is more complex, involving a host of cellular and subcellular changes that impact on neuronal function and viability while also affecting vascular reactivity and the activation of multiple biological response pathways. Here we review the brain's response to injury, examining both focal and diffuse changes and their implications for post-traumatic brain dysfunction and recovery. TBI-induced neuronal dysfunction and death as well as the diffuse involvement of multiple fiber projections are discussed together with considerations of how local axonal membrane changes or channelopathy translate into local ionic dysregulation and axonal disconnection. Concomitant changes in the cerebral microcirculation are also discussed and their relationship with the parallel changes in the brain's metabolism is considered. These cellular and subcellular events occurring within neurons and their blood supply are correlated with multiple biological response modifiers evoked by generalized post-traumatic inflammation and the parallel activation of oxidative stress processes. The chapter closes with considerations of recovery following focal or diffuse injury. Evidence for dynamic brain reorganization/repair is presented, with considerations of traumatically induced circuit disruption and their progression to either adaptive or in some cases, maladaptive reorganization.
Collapse
Affiliation(s)
- Melissa J McGinn
- Department of Anatomy and Neurobiology, Medical College of Virginia Campus of Virginia Commonwealth University, Richmond, VA, USA
| | - John T Povlishock
- Department of Anatomy and Neurobiology, Medical College of Virginia Campus of Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
20
|
Do metals that translocate to the brain exacerbate traumatic brain injury? Med Hypotheses 2014; 82:558-62. [DOI: 10.1016/j.mehy.2014.02.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 01/22/2014] [Accepted: 02/08/2014] [Indexed: 11/19/2022]
|
21
|
Cornelius C, Crupi R, Calabrese V, Graziano A, Milone P, Pennisi G, Radak Z, Calabrese EJ, Cuzzocrea S. Traumatic brain injury: oxidative stress and neuroprotection. Antioxid Redox Signal 2013; 19:836-53. [PMID: 23547621 DOI: 10.1089/ars.2012.4981] [Citation(s) in RCA: 243] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
SIGNIFICANCE A vast amount of circumstantial evidence implicates high energy oxidants and oxidative stress as mediators of secondary damage associated with traumatic brain injury. The excessive production of reactive oxygen species due to excitotoxicity and exhaustion of the endogenous antioxidant system induces peroxidation of cellular and vascular structures, protein oxidation, cleavage of DNA, and inhibition of the mitochondrial electron transport chain. RECENT ADVANCES Different integrated responses exist in the brain to detect oxidative stress, which is controlled by several genes termed vitagens. Vitagens encode for cytoprotective heat shock proteins, and thioredoxin and sirtuins. CRITICAL ISSUES AND FUTURE DIRECTIONS This article discusses selected aspects of secondary brain injury after trauma and outlines key mechanisms associated with toxicity, oxidative stress, inflammation, and necrosis. Finally, this review discusses the role of different oxidants and presents potential clinically relevant molecular targets that could be harnessed to treat secondary injury associated with brain trauma.
Collapse
Affiliation(s)
- Carolin Cornelius
- Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina, Messina, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Resveratrol decreases inflammation in the brain of mice with mild traumatic brain injury. J Trauma Acute Care Surg 2013; 74:470-4; discussion 474-5. [PMID: 23354240 DOI: 10.1097/ta.0b013e31827e1f51] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Following a mild traumatic brain injury (TBI) event, the secondary brain injury that persists after the initial blow to the head consists of excitotoxicity, decreased cerebral glucose levels, oxidant injury, mitochondrial dysfunction, inflammation, and neuronal cell death. To date, there are no effective interventions used at decreasing secondary brain injury after mild TBI. METHODS In this study, male mice were treated with either placebo or resveratrol (100 mg/kg) at 5 minutes and 12 hours after mild TBI. The mice were injured using the controlled cortical impact device. In this closed-head model, a midline incision was made to access the skull and the impactor tip was aligned on the sagittal suture midway between the bregma and lambda sutures. The mice were injured at a depth of 2.0 mm, velocity of 4 m/s, and a delay time of 100 milliseconds. At 72 hours following injury, the animals were intracardially perfused with 0.9% saline followed by 10% phosphate-buffered formalin. The whole brain was removed, sliced, and stained for microglial activation (Iba1). In addition, using the enzyme-linked immunosorbent assay, tissue levels of interleukin 6 (IL-6) and IL-12 were measured in the cerebral cortex and hippocampus. RESULTS In this study, we found that in the placebo treatment group, there was a significant increase in Iba1 staining in the brain. The levels of microglial activation was reduced by resveratrol in the cerebral cortex (p < 0.001), corpus callosum (p < 0.001), and dentate gyrus (p < 0.005) brain regions after mild TBI. In addition to Iba1, resveratrol decreased the brain levels of IL-6 (p < 0.0001) and IL-12 (p < 0.004), which were observed in the hippocampus of the placebo group. In our model, no increase of IL-6 or IL-12 was observed in the cerebral cortex following TBI. CONCLUSION Resveratrol given acutely after TBI results in a decrease in neuroinflammation. These results suggest that resveratrol may be beneficial in reducing secondary brain injury after experiencing a mild TBI.
Collapse
|
23
|
Cervini-Silva J, Nieto-Camacho A, Gomez-Vidales V, Ramírez-Apán MT. Oxidative stress induced by arsenopyrite and the role of desferrioxamine-B as radical scavenger. CHEMOSPHERE 2013; 90:1779-1784. [PMID: 22955046 DOI: 10.1016/j.chemosphere.2012.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2012] [Revised: 07/30/2012] [Accepted: 08/03/2012] [Indexed: 06/01/2023]
Abstract
Arsenopyrite (FeAsS) is one of the earth's primary mineral sources of As, yet its effects on cell damage remain largely unknown. This paper addresses the question whether FeAsS induces lipid peroxidation (LP), a major indicator of oxidative stress. Screening and monitoring of LP was conducted using Thiobarbituric Acid Reactive Substances (TBARSs) assay. The lipid source was supernatant of rat brain homogenates. The formation of TBARS by FeAsS was rapid and took place just after 10 min. Maximum TBARS levels (ca. 14 nmol TBARS per mg of protein) were observed after 1h and remained constant thereafter. Suspension fraction separations showed that dissolved and structural components contributed to LP. The formation of TBARS by soluble As, As(III) or As(V), compared to basal levels. The initiation of LP by FeAsS was consistent with a mechanism initiated by the Fe(3+)/O(2)(-) redox system, and differed initiated by Fe(2+)/O(2). The effectiveness of FeAsS and FeSO(4) as inducer compared, and surpassed that of AAPH. On the other hand, the initiation of LP by FeAsS is consistent with a mechanism initiated by perferryl ion and Fe(3+)/O(2)(-), and differs from the mechanism characteristic of FeSO(4) initiated by the Fe(2+)/O(2) redox system. Proposedly, FeAsS surfaces contain a mixture of Fe(3+) and Fe(2+) that, along with O(2) and O(2)(-), participate in multiple mechanisms of electron transfer. EPR determinations show decreases in DMPO-OH adduct signal in FeAsS suspensions after adding desferrioxamine-B (DFO-B), consistent with the idea that DFO-B serves as a radical scavenger.
Collapse
Affiliation(s)
- Javiera Cervini-Silva
- Departamento de Procesos y Tecnología, Universidad Autónoma Metropolitana Unidad Cuajimalpa, Coyoacán 04510, México DF, Mexico.
| | | | | | | |
Collapse
|
24
|
Granados-Oliveros G, Gómez-Vidales V, Nieto-Camacho A, Morales-Serna JA, Cárdenas J, Salmón M. Photoproduction of H2O2and hydroxyl radicals catalysed by natural and super acid-modified montmorillonite and its oxidative role in the peroxidation of lipids. RSC Adv 2013. [DOI: 10.1039/c2ra22393g] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
25
|
Floyd RA, Towner RA, He T, Hensley K, Maples KR. Translational research involving oxidative stress and diseases of aging. Free Radic Biol Med 2011; 51:931-41. [PMID: 21549833 PMCID: PMC3156308 DOI: 10.1016/j.freeradbiomed.2011.04.014] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Revised: 02/28/2011] [Accepted: 04/07/2011] [Indexed: 01/01/2023]
Abstract
There is ample mounting evidence that reactive oxidant species are exacerbated in inflammatory processes, many pathological conditions, and underlying processes of chronic age-related diseases. Therefore there is increased expectation that therapeutics can be developed that act in some fashion to suppress reactive oxidant species and ameliorate the condition. This has turned out to be more difficult than at first expected. Developing therapeutics for indications in which reactive oxidant species are an important consideration presents some unique challenges. We discuss important questions including whether reactive oxidant species should be a therapeutic target, the need to recognize the fact that an antioxidant in a defined chemical system may be a poor antioxidant operationally in a biological system, and the importance of considering that reactive oxidant species may accompany the disease or pathological system rather than being a causative factor. We also discuss the value of having preclinical models to determine if the processes that are important in causing the disease under study are critically dependent on reactive oxidant species events and if the therapeutic under consideration quells these processes. In addition we discuss measures of success that must be met in commercial research and development and in preclinical and clinical trials and discuss as examples our translational research effort in developing nitrones for the treatment of acute ischemic stroke and as anti-cancer agents.
Collapse
Affiliation(s)
- Robert A Floyd
- Experimental Therapeutics, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.
| | | | | | | | | |
Collapse
|
26
|
Cornejo-Garrido H, Kibanova D, Nieto-Camacho A, Guzmán J, Ramírez-Apan T, Fernández-Lomelín P, Garduño ML, Cervini-Silva J. Oxidative stress, cytoxicity, and cell mortality induced by nano-sized lead in aqueous suspensions. CHEMOSPHERE 2011; 84:1329-1335. [PMID: 21640370 DOI: 10.1016/j.chemosphere.2011.05.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2011] [Revised: 05/04/2011] [Accepted: 05/07/2011] [Indexed: 05/30/2023]
Abstract
This paper reports on the effect of aqueous and nano-particulated Pb on oxidative stress (lipid peroxidation), cytoxicity, and cell mortality. As determined by the Thiobarbituric Acid Reactive Substances (TBARS) method, only 6h after incubation aqueous suspensions bearing nano-sized PbO(2), soluble Pb(II), and brain-homogenate only suspensions, were determined to contain as much as ca. 7, 5, and 1 nmol TBARS mg protein(-1), respectively. Exposure of human cells (central nervous system, prostate, leukemia, colon, breast, lung cells) to nano-PbO(2) led to cell-growth inhibition values (%) ca. ≤18.7%. Finally, as estimated by the Artemia salina test, cell mortality values were found to show high-survival larvae rates. Microscopic observations revealed that Pb particles were swallowed, but caused no mortality, however.
Collapse
Affiliation(s)
- Hilda Cornejo-Garrido
- Departamento de Procesos y Tecnología, Universidad Autónoma Metropolitana Unidad Cuajimalpa, Mexico
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Alfieri A, Srivastava S, Siow RCM, Modo M, Fraser PA, Mann GE. Targeting the Nrf2-Keap1 antioxidant defence pathway for neurovascular protection in stroke. J Physiol 2011; 589:4125-36. [PMID: 21646410 DOI: 10.1113/jphysiol.2011.210294] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Endogenous defence mechanisms by which the brain protects itself against noxious stimuli and recovers from ischaemic damage are a key target of stroke research. The loss of viable brain tissue in the ischaemic core region after stroke is associated with damage to the surrounding area known as the penumbra. Activation of the redox-sensitive transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) plays a pivotal role in the cellular defence against oxidative stress via transcriptional upregulation of phase II defence enzymes and antioxidant stress proteins. Although recent evidence implicates Nrf2 in neuroprotection, it is not known whether activation of this pathway within the neurovascular unit protects the brain against blood-brain barrier breakdown and cerebrovascular inflammation. Targeting the neurovascular unit should provide novel insights for effective treatment strategies and facilitate translation of experimental findings into clinical therapy. This review focuses on the cytoprotective role of Nrf2 in stroke and examines the evidence that the Nrf2-Keap1 defence pathway may serve as a therapeutic target for neurovascular protection.
Collapse
Affiliation(s)
- Alessio Alfieri
- Cardiovascular Division, BHF Centre of Research Excellence, School of Medicine, King's College London, London, UK
| | | | | | | | | | | |
Collapse
|
28
|
Abstract
One of the most investigated molecular mechanisms involved in the secondary pathophysiology of acute spinal cord injury (SCI) is free radical-induced, iron-catalyzed lipid peroxidation (LP) and protein oxidative/nitrative damage to spinal neurons, glia, and microvascular cells. The reactive nitrogen species peroxynitrite and its highly reactive free radicals are key initiators of LP and protein nitration in the injured spinal cord, the biochemistry, and pathophysiology of which are first of all reviewed in this article. This is followed by a presentation of the antioxidant mechanistic approaches and pharmacological compounds that have been shown to have neuroprotective properties in preclinical SCI models. Two of these, which act by inhibition of LP, are high-dose treatment with the glucocorticoid steroid methylprednisolone (MP) and the nonglucocorticoid 21-aminosteroid tirilazad, have been demonstrated in the multicenter NASCIS clinical trials to produce at least a modest improvement in neurological recovery when administered within the first 8 hours after SCI. Although these results have provided considerable validation of oxidative damage as a clinically practical neuroprotective target, there is a need for the discovery of safer and more effective antioxidant compounds for acute SCI.
Collapse
Affiliation(s)
- Edward D Hall
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, Kentucky 40506, USA.
| |
Collapse
|
29
|
Pillai DR, Heidemann RM, Kumar P, Shanbhag N, Lanz T, Dittmar MS, Sandner B, Beier CP, Weidner N, Greenlee MW, Schuierer G, Bogdahn U, Schlachetzki F. Comprehensive small animal imaging strategies on a clinical 3 T dedicated head MR-scanner; adapted methods and sequence protocols in CNS pathologies. PLoS One 2011; 6:e16091. [PMID: 21326876 PMCID: PMC3034718 DOI: 10.1371/journal.pone.0016091] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Accepted: 12/09/2010] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Small animal models of human diseases are an indispensable aspect of pre-clinical research. Being dynamic, most pathologies demand extensive longitudinal monitoring to understand disease mechanisms, drug efficacy and side effects. These considerations often demand the concomitant development of monitoring systems with sufficient temporal and spatial resolution. METHODOLOGY AND RESULTS This study attempts to configure and optimize a clinical 3 Tesla magnetic resonance scanner to facilitate imaging of small animal central nervous system pathologies. The hardware of the scanner was complemented by a custom-built, 4-channel phased array coil system. Extensive modification of standard sequence protocols was carried out based on tissue relaxometric calculations. Proton density differences between the gray and white matter of the rodent spinal cord along with transverse relaxation due to magnetic susceptibility differences at the cortex and striatum of both rats and mice demonstrated statistically significant differences. The employed parallel imaging reconstruction algorithms had distinct properties dependent on the sequence type and in the presence of the contrast agent. The attempt to morphologically phenotype a normal healthy rat brain in multiple planes delineated a number of anatomical regions, and all the clinically relevant sequels following acute cerebral ischemia could be adequately characterized. Changes in blood-brain-barrier permeability following ischemia-reperfusion were also apparent at a later time. Typical characteristics of intra-cerebral haemorrhage at acute and chronic stages were also visualized up to one month. Two models of rodent spinal cord injury were adequately characterized and closely mimicked the results of histological studies. In the employed rodent animal handling system a mouse model of glioblastoma was also studied with unequivocal results. CONCLUSIONS The implemented customizations including extensive sequence protocol modifications resulted in images of high diagnostic quality. These results prove that lack of dedicated animal scanners shouldn't discourage conventional small animal imaging studies.
Collapse
Affiliation(s)
- Deepu R. Pillai
- Department of Neurology, Regensburg University Medical Centre, Regensburg, Germany
- Department of Genetics and Neurobiology, Biozentrum, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Robin M. Heidemann
- Department of Neurophysics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Siemens Healthcare Sector, Erlangen, Germany
| | - Praveen Kumar
- Department of Neurology, Regensburg University Medical Centre, Regensburg, Germany
- Department of Neurology, University Medical Centre, RWTH Aachen, Aachen, Germany
| | - Nagesh Shanbhag
- Department of Neurology, Regensburg University Medical Centre, Regensburg, Germany
| | - Titus Lanz
- RAPID Biomedical GmbH, Würzburg-Rimpar, Germany
| | - Michael S. Dittmar
- Department of Anaesthesiology, Regensburg University Medical Centre, Regensburg, Germany
| | - Beatrice Sandner
- Department of Neurology, Regensburg University Medical Centre, Regensburg, Germany
| | - Christoph P. Beier
- Department of Neurology, Regensburg University Medical Centre, Regensburg, Germany
- Department of Neurology, University Medical Centre, RWTH Aachen, Aachen, Germany
| | - Norbert Weidner
- Department of Neurology, Regensburg University Medical Centre, Regensburg, Germany
- Institute for Paraplegia, University of Heidelberg, Heidelberg, Germany
| | - Mark W. Greenlee
- Institute for Experimental Psychology, University of Regensburg, Regensburg, Germany
| | - Gerhard Schuierer
- Center for Neuroradiology, Regensburg University Medical Centre and Bezirksklinikum Regensburg, Regensburg, Germany
| | - Ulrich Bogdahn
- Department of Neurology, Regensburg University Medical Centre, Regensburg, Germany
| | - Felix Schlachetzki
- Department of Neurology, Regensburg University Medical Centre, Regensburg, Germany
- * E-mail:
| |
Collapse
|
30
|
Lithium and valproate modulate antioxidant enzymes and prevent ouabain-induced oxidative damage in an animal model of mania. J Psychiatr Res 2011; 45:162-8. [PMID: 20627318 DOI: 10.1016/j.jpsychires.2010.05.011] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2010] [Revised: 05/04/2010] [Accepted: 05/07/2010] [Indexed: 10/19/2022]
Abstract
In this study, we assessed the oxidative stress parameters in rats submitted to an animal model of mania induced by ouabain (OUA), which included the use of lithium (Li) and valproate (VPA). Li and VPA treatment reversed and prevented the OUA-induced damage in these structures, however, this effect varies depending on the brain region and treatment regimen. Moreover, the activity of the antioxidant enzymes, namely, superoxide dismutase (SOD) and catalase (CAT) was found to be increased and decreased, respectively, in the brain of OUA-administered rats. Li and VPA modulated SOD and CAT activities in OUA-subjected rats in both experimental models. Our results support the notion that Li and VPA exert antioxidant-like properties in the brain of rats submitted to animal model of mania induced by ouabain.
Collapse
|
31
|
Hadjipavlou-Litina D, Magoulas GE, Bariamis SE, Drainas D, Avgoustakis K, Papaioannou D. Does conjugation of antioxidants improve their antioxidative/anti-inflammatory potential? Bioorg Med Chem 2010; 18:8204-17. [PMID: 21041094 DOI: 10.1016/j.bmc.2010.10.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Revised: 09/30/2010] [Accepted: 10/06/2010] [Indexed: 11/17/2022]
Abstract
A series of symmetric and asymmetric spermine (SPM) conjugates with all-trans-retinoic acid (ATRA), acitretin (ACI), (E)-3-(trioxsalen-4'-yl)acrylic acid (TRAA) and L-DOPA, amides of ACI, l-DOPA and TRAA with 1-aminobutane, benzylamine, dopamine and 1,12-diaminobutane as well as hybrid conjugates of O,O'-dimethylcaffeic acid (DMCA) with TRAA or N-fumaroyl-indole-3-carboxanilide (FICA) and 2-(2-aminoethoxy)ethanol were synthesized and their antioxidant properties were studied. The reducing activity (RA)% of the compounds were evaluated using the 1,1-diphenyl-2-picrylhydrazyl (DPPH) free radical-scavenging assay and found to be in the range 0-92(20 min)%/96(60 min)% at 100μM, the most powerful being the conjugates L-DOPA-SPM-L-DOPA (8, RA=89%/96%) and L-DOPA-dopamine (13, RA=92%/92%). Conjugate DMCA-NH(CH₂CH₂O)₂-FICA (14) was the most powerful LOX inhibitor with IC₅₀ 33.5μM, followed by the conjugates ACI-NHCH₂Ph (10, IC₅₀ 40.5μM), ACI-SPM-TRAA (7, IC₅₀ 41.5μM), DMCA-NH(CH₂CH₂O)₂-TRAA (15, IC₅₀ 65μM), 13 (IC₅₀ 81.5μM) and ACI-dopamine (11, IC₅₀ 87μM). The most potent inhibitors of lipid peroxidation at 100μM were the conjugates 15 (98%) and ACI-SPM-ACI (4, 97%) whereas all other compounds showed activities comparable or lower than trolox. The most interesting compounds, namely ATRA-SPM-ATRA (3), 4, 10, 11 and 15, as well as unconjugated compounds such as ATRA and dopamine, were studied for their anti-inflammatory activity in vivo on rat paw oedema induced by Carrageenan and found to exhibit, for doses of 0.01 mmol/mL of conjugates per Kg of rat body weight, weaker anti-inflammatory activities (3.6-40%) than indomethacin (47%) with conjugate 3 being the most potent (40%) in this series of compounds. The cytocompatibility of selected compounds was evaluated by the viability of RAMEC cells in the presence of different concentrations (0.5-50μM) of the compounds. Conjugates 3 (IC₅₀ 2.6μM) and 4 (IC₅₀ 4.7μM) were more cytotoxic than the corresponding unconjugated retinoids ATRA (IC₅₀ 18.3μM) and ACI (IC₅₀ 14.6μM), whereas conjugate 15 (IC₅₀ 12.9μM) was less cytotoxic than either DCSP (IC₅₀ 11.3μM) or the tert-butyl ester of TRAA (IC₅₀ 2.9μM).
Collapse
|
32
|
Role of oxidative stress in the pathophysiology of bipolar disorder. Neurochem Res 2010; 35:1295-301. [DOI: 10.1007/s11064-010-0195-2] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2010] [Indexed: 11/30/2022]
|
33
|
Hall ED, Vaishnav RA, Mustafa AG. Antioxidant therapies for traumatic brain injury. Neurotherapeutics 2010; 7:51-61. [PMID: 20129497 PMCID: PMC2818465 DOI: 10.1016/j.nurt.2009.10.021] [Citation(s) in RCA: 272] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Accepted: 10/19/2009] [Indexed: 12/31/2022] Open
Abstract
Free radical-induced oxidative damage reactions, and membrane lipid peroxidation (LP), in particular, are among the best validated secondary injury mechanisms in preclinical traumatic brain injury (TBI) models. In addition to the disruption of the membrane phospholipid architecture, LP results in the formation of cytotoxic aldehyde-containing products that bind to cellular proteins and impair their normal functions. This article reviews the progress of the past three decades in regard to the preclinical discovery and attempted clinical development of antioxidant drugs designed to inhibit free radical-induced LP and its neurotoxic consequences via different mechanisms including the O(2)(*-) scavenger superoxide dismutase and the lipid peroxidation inhibitor tirilazad. In addition, various other antioxidant agents that have been shown to have efficacy in preclinical TBI models are briefly presented, such as the LP inhibitors U83836E, resveratrol, curcumin, OPC-14177, and lipoic acid; the iron chelator deferoxamine and the nitroxide-containing antioxidants, such as alpha-phenyl-tert-butyl nitrone and tempol. A relatively new antioxidant mechanistic strategy for acute TBI is aimed at the scavenging of aldehydic LP byproducts that are highly neurotoxic with "carbonyl scavenging" compounds. Finally, it is proposed that the most effective approach to interrupt posttraumatic oxidative brain damage after TBI might involve the combined treatment with mechanistically complementary antioxidants that simultaneously scavenge LP-initiating free radicals, inhibit LP propagation, and lastly remove neurotoxic LP byproducts.
Collapse
Affiliation(s)
- Edward D Hall
- Spinal Cord & Brain Injury Research Center, University of Kentucky Medical Center, Lexington, Kentucky 40536, USA.
| | | | | |
Collapse
|
34
|
Liu Y, Wang CY, Kong XH, Wang HJ, Chang J, Zhang DP, Ban DX, Feng SQ. Novel multifunctional polyethylene glycol-transactivating-transduction protein-modified liposomes cross the blood-spinal cord barrier after spinal cord injury. J Drug Target 2009; 18:420-9. [DOI: 10.3109/10611860903434001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
35
|
Abstract
Multiple sclerosis (MS) is primarily an autoimmune disorder of unknown origin. This review focuses iron overload and oxidative stress as surrounding cause that leads to immunomodulation in chronic MS. Iron overload has been demonstrated in MS lesions, as a feature common with other neurodegenerative disorders. However, the recent description of chronic cerebrospinal venous insufficiency (CCSVI) associated to MS, with significant anomalies in cerebral venous outflow hemodynamics, permit to propose a parallel with chronic venous disorders (CVDs) in the mechanism of iron deposition. Abnormal cerebral venous reflux is peculiar to MS, and was not found in a miscellaneous of patients affected by other neurodegenerative disorders characterized by iron stores, such as Parkinson's, Alzheimer's, amyotrophic lateral sclerosis. Several recently published studies support the hypothesis that MS progresses along the venous vasculature. The peculiarity of CCSVI-related cerebral venous blood flow disturbances, together with the histology of the perivenous spaces and recent findings from advanced magnetic resonance imaging techniques, support the hypothesis that iron deposits in MS are a consequence of altered cerebral venous return and chronic insufficient venous drainage.
Collapse
|
36
|
Involvement of extracellular ascorbate and iron in hydroxyl radical generation in rat striatum in carbon monoxide poisoning. Toxicology 2009; 264:69-73. [DOI: 10.1016/j.tox.2009.07.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2009] [Revised: 07/07/2009] [Accepted: 07/17/2009] [Indexed: 11/20/2022]
|
37
|
Floyd RA. Serendipitous findings while researching oxygen free radicals. Free Radic Biol Med 2009; 46:1004-13. [PMID: 19439210 PMCID: PMC2683184 DOI: 10.1016/j.freeradbiomed.2009.02.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2009] [Revised: 02/05/2009] [Accepted: 02/05/2009] [Indexed: 11/13/2022]
Abstract
This review is based on the honor of receiving the Discovery Award from the Society of Free Radical Biology and Medicine. The review is reflective and presents our thinking that led to experiments that yielded novel observations. Critical questioning of our understanding of oxygen free radicals in biomedical problems led us to use and develop more direct and extremely sensitive methods. This included nitrone free radical spin trapping and HPLC-electrochemical detection. This technology led to the pioneering use of salicylate to trap hydroxyl free radicals and show increased flux in ischemia/reperfused brain regions and also to first sensitively detect 8-hydroxyl-2-deoxyguanosine in oxidatively damaged DNA and help assess its role in cancer development. We demonstrated that methylene blue (MB) photoinduces formation of 8-hydroxyguanine in DNA and RNA and discovered that MB sensitively photoinactivates RNA viruses, including HIV and the West Nile virus. Studies in experimental stroke led us serendipitously to discover that alpha-phenyl-tert-butylnitrone (PBN) was neuroprotective if given after the stroke. This led to extensive commercial development of NXY-059, a PBN derivative, for the treatment of stroke. More recently we discovered that PBN nitrones have potent anti-cancer activity and are active in preventing hearing loss caused by acute acoustical trauma.
Collapse
Affiliation(s)
- Robert A Floyd
- Experimental Therapeutics Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73170, USA.
| |
Collapse
|
38
|
Leipnitz G, Seminotti B, Fernandes CG, Amaral AU, Beskow AP, Silva LDB, Zanatta Â, Ribeiro CA, Vargas CR, Wajner M. Striatum is more vulnerable to oxidative damage induced by the metabolites accumulating in 3‐hydroxy‐3‐methylglutaryl‐CoA lyase deficiency as compared to liver. Int J Dev Neurosci 2009; 27:351-6. [DOI: 10.1016/j.ijdevneu.2009.03.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2009] [Revised: 02/11/2009] [Accepted: 03/03/2009] [Indexed: 01/20/2023] Open
Affiliation(s)
- Guilhian Leipnitz
- Departamento de BioquímicaInstituto de Ciências Básicas da SaúdeUFRGSPorto AlegreRSBrazil
| | - Bianca Seminotti
- Departamento de BioquímicaInstituto de Ciências Básicas da SaúdeUFRGSPorto AlegreRSBrazil
| | - Carolina G. Fernandes
- Departamento de BioquímicaInstituto de Ciências Básicas da SaúdeUFRGSPorto AlegreRSBrazil
| | - Alexandre U. Amaral
- Departamento de BioquímicaInstituto de Ciências Básicas da SaúdeUFRGSPorto AlegreRSBrazil
| | - Ana Paula Beskow
- Departamento de BioquímicaInstituto de Ciências Básicas da SaúdeUFRGSPorto AlegreRSBrazil
| | - Lucila de B. Silva
- Departamento de BioquímicaInstituto de Ciências Básicas da SaúdeUFRGSPorto AlegreRSBrazil
| | - Ângela Zanatta
- Departamento de BioquímicaInstituto de Ciências Básicas da SaúdeUFRGSPorto AlegreRSBrazil
| | - César A.J. Ribeiro
- Departamento de BioquímicaInstituto de Ciências Básicas da SaúdeUFRGSPorto AlegreRSBrazil
| | - Carmen R. Vargas
- Departamento de BioquímicaInstituto de Ciências Básicas da SaúdeUFRGSPorto AlegreRSBrazil
| | - Moacir Wajner
- Departamento de BioquímicaInstituto de Ciências Básicas da SaúdeUFRGSPorto AlegreRSBrazil
- Serviço de Genética MédicaHospital de Clínicas de Porto AlegreRSBrazil
- Universidade Luterana do BrasilCanoasRSBrazil
| |
Collapse
|
39
|
Uttara B, Singh AV, Zamboni P, Mahajan R. Oxidative stress and neurodegenerative diseases: a review of upstream and downstream antioxidant therapeutic options. Curr Neuropharmacol 2009; 7:65-74. [PMID: 19721819 PMCID: PMC2724665 DOI: 10.2174/157015909787602823] [Citation(s) in RCA: 2122] [Impact Index Per Article: 141.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2008] [Revised: 11/19/2008] [Accepted: 11/28/2008] [Indexed: 02/06/2023] Open
Abstract
Free radicals are common outcome of normal aerobic cellular metabolism. In-built antioxidant system of body plays its decisive role in prevention of any loss due to free radicals. However, imbalanced defense mechanism of antioxidants, overproduction or incorporation of free radicals from environment to living system leads to serious penalty leading to neuro-degeneration. Neural cells suffer functional or sensory loss in neurodegenerative diseases. Apart from several other environmental or genetic factors, oxidative stress (OS) leading to free radical attack on neural cells contributes calamitous role to neuro-degeneration. Though, oxygen is imperative for life, imbalanced metabolism and excess reactive oxygen species (ROS) generation end into a range of disorders such as Alzheimer's disease, Parkinson's disease, aging and many other neural disorders. Toxicity of free radicals contributes to proteins and DNA injury, inflammation, tissue damage and subsequent cellular apoptosis. Antioxidants are now being looked upon as persuasive therapeutic against solemn neuronal loss, as they have capability to combat by neutralizing free radicals. Diet is major source of antioxidants, as well as medicinal herbs are catching attention to be commercial source of antioxidants at present. Recognition of upstream and downstream antioxidant therapy to oxidative stress has been proved an effective tool in alteration of any neuronal damage as well as free radical scavenging. Antioxidants have a wide scope to sequester metal ions involved in neuronal plaque formation to prevent oxidative stress. In addition, antioxidant therapy is vital in scavenging free radicals and ROS preventing neuronal degeneration in post-oxidative stress scenario.
Collapse
Affiliation(s)
- Bayani Uttara
- Department of Biotechnology, M. J. College, M. J. Road, Jalgaon- 425 001, India
| | - Ajay V. Singh
- Centro Interdisciplinare Materiali e Interfacce Nanostrutturati (CIMAINA), Dipartimento di Fisica, Universita di Milano, Via Celoria 16, 20133 Milan, Italy
- Centre for Vascular Disease, University of Ferrara, 41100 Ferrara, Italy
| | - Paolo Zamboni
- Centre for Vascular Disease, University of Ferrara, 41100 Ferrara, Italy
| | - R.T Mahajan
- Department of Biotechnology, M. J. College, M. J. Road, Jalgaon- 425 001, India
| |
Collapse
|
40
|
Manda K, Ueno M, Anzai K. Space radiation-induced inhibition of neurogenesis in the hippocampal dentate gyrus and memory impairment in mice: ameliorative potential of the melatonin metabolite, AFMK. J Pineal Res 2008; 45:430-8. [PMID: 18631288 DOI: 10.1111/j.1600-079x.2008.00611.x] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Evaluation of potential health effects from high energy charged particle radiation exposure during long duration space travel is important for the future of manned missions. Cognitive health of an organism is considered to be maintained by the capacity of hippocampal precursors to proliferate and differentiate. Environmental stressors including irradiation have been shown to inhibit neurogenesis and are associated with the onset of cognitive impairments. The present study reports on the protective effects of N(1)-acetyl-N(2)-formyl-5-methoxykynuramine (AFMK), a melatonin metabolite, against high energy charged particle radiation-induced oxidative damage to the brain. We observed that radiation exposure (2.0 Gy of 500 MeV/nucleon (56)Fe beams, a ground-based model of space radiation) impaired the spatial memory of mice at later intervals without affecting the motor activities. AFMK pretreatment significantly ameliorated these neurobehavioral ailments. Radiation-induced changes in the population of immature and proliferating neurons in the dentate gyrus were localized using anti-doublecortin (Dcx) and anti-Ki-67 expression. AFMK pretreatment significantly inhibited the loss of Dcx and Ki-67 positive cells. Moreover, AFMK pretreatment ameliorated the radiation-induced augmentation of protein carbonyls and 4-hydroxyalkenal + malondialdehyde (MDA + HAE) in the brain and maintained the total antioxidant capacity of plasma and nonprotein sulfhydryl contents in brain.
Collapse
Affiliation(s)
- Kailash Manda
- National Institute of Radiological Science, Chiba, Japan.
| | | | | |
Collapse
|
41
|
Heo HJ, Choi SJ, Choi SG, Shin DH, Lee JM, Lee CY. Effects of banana, orange, and apple on oxidative stress-induced neurotoxicity in PC12 cells. J Food Sci 2008; 73:H28-32. [PMID: 18298733 DOI: 10.1111/j.1750-3841.2007.00632.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Banana, orange, and apple are the major fruits in Western and Asian diets. In order to find the effects of these fruits, neuron like PC12 cells were exposed to the extracts of these fruits before H(2)O(2) treatment. We found a significant viability of PC12 cells by the MTT reduction test, which indicated that the phenolics of banana, orange, and apple fruits prevented oxidative stress-induced neurotoxicity. Additional tests by lactate dehydrogenase and trypan blue exclusion assays showed that the extracts reduced oxidative stress-induced neuronal cell membrane damage. These results suggest that fresh apples, banana, and orange in our daily diet along with other fruits may protect neuron cells against oxidative stress-induced neurotoxicity and may play an important role in reducing the risk of neurodegenerative disorders such as Alzheimer's disease.
Collapse
Affiliation(s)
- H J Heo
- Div. of Applied Life Science, Inst. of Agriculture and Life Sciences, Gyeongsang Natl. Univ., Jinju, Gyeongnam 660-701, Korea
| | | | | | | | | | | |
Collapse
|
42
|
Silva-Adaya D, Pérez-De La Cruz V, Herrera-Mundo MN, Mendoza-Macedo K, Villeda-Hernández J, Binienda Z, Ali SF, Santamaría A. Excitotoxic damage, disrupted energy metabolism, and oxidative stress in the rat brain: antioxidant and neuroprotective effects of L-carnitine. J Neurochem 2008; 105:677-89. [PMID: 18194214 DOI: 10.1111/j.1471-4159.2007.05174.x] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Excitotoxicity and disrupted energy metabolism are major events leading to nerve cell death in neurodegenerative disorders. These cooperative pathways share one common aspect: triggering of oxidative stress by free radical formation. In this work, we evaluated the effects of the antioxidant and energy precursor, levocarnitine (L-CAR), on the oxidative damage and the behavioral, morphological, and neurochemical alterations produced in nerve tissue by the excitotoxin and free radical precursor, quinolinic acid (2,3-pyrindin dicarboxylic acid; QUIN), and the mitochondrial toxin, 3-nitropropionic acid (3-NP). Oxidative damage was assessed by the estimation of reactive oxygen species formation, lipid peroxidation, and mitochondrial dysfunction in synaptosomal fractions. Behavioral, morphological, and neurochemical alterations were evaluated as markers of neurotoxicity in animals systemically administered with L-CAR, chronically injected with 3-NP and/or intrastriatally infused with QUIN. At micromolar concentrations, L-CAR reduced the three markers of oxidative stress stimulated by both toxins alone or in combination. L-CAR also prevented the rotation behavior evoked by QUIN and the hypokinetic pattern induced by 3-NP in rats. Morphological alterations produced by both toxins (increased striatal glial fibrillary acidic protein-immunoreactivity for QUIN and enhanced neuronal damage in different brain regions for 3-NP) were reduced by L-CAR. In addition, L-CAR prevented the synergistic action of 3-NP and QUIN to increase motor asymmetry and depleted striatal GABA levels. Our results suggest that the protective properties of L-CAR in the neurotoxic models tested are mostly mediated by its characteristics as an antioxidant agent.
Collapse
Affiliation(s)
- Daniela Silva-Adaya
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, México, Mexico
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Latini A, Scussiato K, Leipnitz G, Gibson KM, Wajner M. Evidence for oxidative stress in tissues derived from succinate semialdehyde dehydrogenase-deficient mice. J Inherit Metab Dis 2007; 30:800-10. [PMID: 17885820 DOI: 10.1007/s10545-007-0599-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2007] [Revised: 04/19/2007] [Accepted: 06/08/2007] [Indexed: 02/06/2023]
Abstract
Animal models of inborn errors of metabolism are useful for investigating the pathogenesis associated with the corresponding human disease. Since the mechanisms involved in the pathophysiology of succinate semialdehyde dehydrogenase (SSADH) deficiency (Aldh5a1; OMIM 271980) are still not established, in the present study we evaluated the tissue antioxidant defences and lipid peroxidation in various cerebral structures (cortex, cerebellum, thalamus and hippocampus) and in the liver of SSADH-deficient mice. The parameters analysed were total radical-trapping antioxidant potential (TRAP) and glutathione (GSH) levels, the activities of the antioxidant enzymes superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPx), as well as thiobarbituric acid-reactive substances (TBARS). We first observed that the tissue nonenzymatic antioxidant defences were significantly reduced in the SSADH-deficient animals, particularly in the liver (decreased TRAP and GSH) and in the cerebral cortex (decreased GSH), as compared to the wild-type mice. Furthermore, SOD activity was significantly increased in the liver and cerebellum, whereas the activity of CAT was significantly higher in the thalamus. In contrast, GPx activity was significantly diminished in the hippocampus. Finally, we observed that lipid peroxidation (TBARS levels) was markedly increased in the liver and cerebral cortex, reflecting a high lipid oxidative damage in these tissues. Our data showing an imbalance between tissue antioxidant defences and oxidative attack strongly indicate that oxidative stress is involved in the pathophysiology of SSADH deficiency in mice, and likely the corresponding human disorder.
Collapse
Affiliation(s)
- A Latini
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | | | | | | | | |
Collapse
|
44
|
Choi JH, Kim DH, Yun IJ, Chang JH, Chun BG, Choi SH. Zaprinast inhibits hydrogen peroxide-induced lysosomal destabilization and cell death in astrocytes. Eur J Pharmacol 2007; 571:106-15. [PMID: 17643412 DOI: 10.1016/j.ejphar.2007.06.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2006] [Revised: 06/05/2007] [Accepted: 06/07/2007] [Indexed: 12/25/2022]
Abstract
The lysosomal destabilization that precedes mitochondrial apoptotic changes is an important step in cell death, particularly in oxidative cell death. This study describes the novel pharmacological effects of zaprinast, a cGMP-elevating phosphodiesterase inhibitor, on the inhibition of oxidative cell death in astrocyte cultures. H2O2-induced oxidative cytotoxicity was measured grossly by monitoring lactate dehydrogenase (LDH) release, and was found to be associated with lysosomal acridine orange relocation, lysosomal cathepsin D release into cytosol, and reduced mitochondrial potentials. Moreover, zaprinast (100 microM) inhibited all of these cytotoxic phenomena. In addition, H2O2-induced LDH release was not inhibited by 8-pCPT-cGMP, and the inhibition of this release by zaprinast was unaffected by Rp-8-pCPT-cGMP, a protein kinase G inhibitor. Zaprinast was found to inhibit sphingosine-induced lysosomal acridine orange relocation and the induced decrease in mitochondrial potential, but zaprinast had no effect on rotenone-induced mitochondrial collapse, which was not associated with lysosomal destabilization. However, zaprinast did not inhibit the cellular increase of reactive oxygen species induced by H2O2, which suggests that its protective mechanism differs from that of desferrioxamine, which does inhibit such cellular increase of oxygen free radicals. We suggest that the novel protective effect of zaprinast on H2O2-induced oxidative cell death is primarily associated with its inhibition of lysosomal destabilization.
Collapse
Affiliation(s)
- Jae-Hyuck Choi
- Department of Pharmacology, Korea University College of Medicine, 126-1, 5-Ga, Anam-Dong, Sungbuk-Gu, Seoul 136-705, Republic of Korea
| | | | | | | | | | | |
Collapse
|
45
|
Schonfeld E, Yasharel I, Yavin E, Brand A. Docosahexaenoic Acid Enhances Iron Uptake by Modulating Iron Transporters and Accelerates Apoptotic Death in PC12 Cells. Neurochem Res 2007; 32:1673-84. [PMID: 17551831 DOI: 10.1007/s11064-007-9378-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2007] [Accepted: 05/08/2007] [Indexed: 01/18/2023]
Abstract
The effect of docosahexaenoic acid (DHA; 22:6 n-3) on Fe(2+)-mediated and/or H(2)O(2)-mediated oxidative stress (OS) was investigated in a PC12 pheochromocytoma cell line in the presence or absence of 50 ng/ml nerve growth factor (NGF). DHA-supplemented cells showed enhanced Fe(2+)-induced cell damage as evident by increased lipid peroxides formation (10-fold) and reduced neutral red (NR) dye uptake in a NGF-independent fashion. DHA caused a nearly 10-fold increase in free iron uptake in NGF-treated cells and doubled iron uptake in nondifferentiated cells. DHA-enrichment induced an elevation in the transferrin receptor protein in the nondifferentiated cells whereas NGF-treatment led to a substantial increase in the ubiquitous divalent metal ion transporter 1 (DMT-1) as detected by mRNA levels using qRT-PCR. The mechanism of action of DHA to accelerate cell death may be associated with the externalization of amino-phosphoglycerides (PG) species of which, increased ethanolamine plasmalogen levels, may be essential for cell rescue as noted in NGF-treated PC12 cells.
Collapse
Affiliation(s)
- Eldi Schonfeld
- Department of Neurobiology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | | | | | | |
Collapse
|
46
|
Kaneko N, Sugioka T, Sakurai H. Aluminum compounds enhance lipid peroxidation in liposomes: Insight into cellular damage caused by oxidative stress. J Inorg Biochem 2007; 101:967-75. [PMID: 17467804 DOI: 10.1016/j.jinorgbio.2007.03.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2007] [Revised: 03/09/2007] [Accepted: 03/14/2007] [Indexed: 11/24/2022]
Abstract
Aluminum (Al) has been proposed as one of the critical environmental factors responsible for several neurodegenerative diseases such as Alzheimer's disease. However, the suggested mechanism involving the contribution of reactive oxygen species still remains controversial. We have first attempted to identify Al compounds either in its ionic or complexed forms that cause oxidative stress in biological systems. For this purpose, we examined the effect of inorganic Fe(2+)- and organic radical initiator (2,2'-azobis (2-amidinopopane) hydrochloride; AAPH)-induced lipid peroxidation by using aluminum (Al(3+)) nitrate and tris(maltolato)aluminum(III) complex (ALM) with respect to molecular oxygen (O(2)) consumption and membrane fluidity change in liposomes as biological membrane models. The following important results were obtained: (1) ALM enhanced the lipid peroxidation induced by Fe(2+) and AAPH in phosphatidylcholine liposomes; this corresponded well with the promotion of O(2) uptake in the same liposomes, (2) Al(3+) increased both lipid peroxidation and O(2) consumption in phosphatidylserine liposomes in the presence of Fe(2+), and (3) both Al(3+) and ALM affected the membrane fluidity on the inner side. It has been concluded that ALM induces higher lipid peroxidation in liposomes than Al(3+); this finding will be useful to gain an insight into the role of Al in cellular damage in relation to oxidative stress.
Collapse
Affiliation(s)
- Noritsugu Kaneko
- Department of Analytical and Bioinorganic Chemistry, Kyoto Pharmaceutical University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan
| | | | | |
Collapse
|
47
|
Manda K, Ueno M, Anzai K. AFMK, a melatonin metabolite, attenuates X-ray-induced oxidative damage to DNA, proteins and lipids in mice. J Pineal Res 2007; 42:386-93. [PMID: 17439555 DOI: 10.1111/j.1600-079x.2007.00432.x] [Citation(s) in RCA: 174] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Antioxidant function of melatonin is well established. However, N(1)-acetyl-N(2)-formyl-5-methoxykynuramine (AFMK), a melatonin metabolite is a sparingly investigated biogenic amine, especially in relation to its in vivo antioxidant function. We have evaluated the oxidative damage to biomolecules (DNA, protein and lipid) induced by X-irradiation in C57BL mice and the prophylactic action of AFMK. The extent of DNA damage was analyzed by single-cell gel electrophoresis in cerebral cortex and serum 8-hydroxydeoxyguanosine (8-OHdG) levels by enzyme-linked immunosorbent assay. Oxidative modification of protein and lipid was measured in the terms of carbonyl content and 4-HAE + MDA (4-hydroxyalkenal + malondialdehyde) status of brain cortex. Radiation exposure dramatically augmented the level of 8-OHdG in serum as well as DNA migration in the comet tail. AFMK pretreatment significantly inhibited DNA damage. In addition, radiation-induced augmentation of protein carbonyl content and HAE + MDA was ameliorated by AFMK pretreatment. Whole-body exposure of mice to X-irradiation also reduced the level of brain sulfhydryl contents (protein-bound sulfhydryl, total sulfhydryl, and nonprotein sulfhydryl) which were significantly protected by AFMK. Radiation-induced decline in the total antioxidant capacity of plasma was significantly reversed in AFMK pretreated mice. Moreover, AFMK showed a very high level of in vitro hydroxyl radical scavenging potential which was measured by an electron spin resonance (ESR) study of the 2-hydroxy-5,5-dimethyl-1-pyrrolineN-oxide (DMPO-OH) adduct. IC(50) values resulting from ESR analysis was 338.08 nm. The present study indicate that AFMK is a potent antioxidant in both in vivo and in vitro systems.
Collapse
Affiliation(s)
- Kailash Manda
- National Institute of Radiological Science, Chiba, Japan
| | | | | |
Collapse
|
48
|
Manda K, Ueno M, Moritake T, Anzai K. Radiation-induced cognitive dysfunction and cerebellar oxidative stress in mice: Protective effect of α-lipoic acid. Behav Brain Res 2007; 177:7-14. [PMID: 17145083 DOI: 10.1016/j.bbr.2006.11.013] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2006] [Revised: 11/06/2006] [Accepted: 11/09/2006] [Indexed: 10/23/2022]
Abstract
Reactive oxygen species are implicated in neurodegeneration and cognitive disorders due to higher vulnerability of neuronal tissues. The cerebellum is recently reported to be involved in cognitive function. Therefore, present study aimed at investigating the role alpha-lipoic acid against radiation-induced oxidative stress and antioxidant status in cerebellum and its correlation with cognitive dysfunction. We observed spontaneous motor activities and spatial memory task of mice using pyroelectric infrared sensor and programmed video tracking system, respectively. Whole body X-irradiation (6 Gy) of mice substantially impaired the reference memory and motor activities of mice. However, acute intraperitoneal treatment of mice with alpha-lipoic acid prior to irradiation significantly attenuated such cognitive dysfunction. Alpha-lipoic acid pretreatment exerted a very high magnitude of protection against radiation-induced augmentation of protein carbonyls and thiobarbituric acid reactive substance (TBARS) in mice cerebellum. Further, radiation-induced deficit of total, nonprotein and protein-bound sulfhydryl (T-SH, NP-SH, PB-SH) contents of cerebellum and plasma ferric reducing power (FRAP) was also inhibited by alpha-lipoic acid pre-treatment. Moreover, alpha-lipoic acid treated mice showed an intact cytoarchitecture of cerebellum, higher counts of intact Purkinje cells and granular cells in comparison to untreated irradiated mice. Results clearly indicate that alpha-lipoic acid is potent neuroprotective antioxidant.
Collapse
Affiliation(s)
- Kailash Manda
- National Institute of Radiological Sciences, Chiba-shi-263-8555, Japan.
| | | | | | | |
Collapse
|
49
|
Jafari M. Dose- and time-dependent effects of sulfur mustard on antioxidant system in liver and brain of rat. Toxicology 2007; 231:30-9. [PMID: 17222496 DOI: 10.1016/j.tox.2006.11.048] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2006] [Revised: 11/07/2006] [Accepted: 11/08/2006] [Indexed: 11/29/2022]
Abstract
This study investigates the dose- and time-dependent effects of sulfur mustard (SM) on antioxidant system and lipid peroxidation in liver and brain of rats. For this purpose, male Wistar rats were randomly divided into eight groups and treated as follows: group 1 as control and groups 2-8 as experimental groups that received SM (1-80 mg/kg) through intraperitoneal injection. Rats were killed after 2, 7 and 14 days of exposure. SM dose-dependently decreased body weight. Superoxide dismutase (SOD), catalase (CAT) and glutathione S-transferase (GST) activities in liver were significantly increased at SM doses lower than 10 mg/kg after 2 and 7 days of exposure. However, the recovery of these parameters was observed after 14 days. At these concentrations, no significant change in glutathione (GSH) and malondialdehyde (MDA) levels were observed. At doses higher than 10 mg/kg, SM significantly decreased SOD, CAT, glutathione peroxidase (GPX), and GST activities in liver and brain and decreased glutathione reductase (GR) activity in liver, which was associated with a depletion of GSH and increased MDA level. Present data indicate that the effect of SM is dose- and time-dependent and at higher doses (>10 mg/kg) induces an oxidative stress response by depleting the antioxidant defense systems and increasing lipid peroxidation in liver and brain of rats.
Collapse
Affiliation(s)
- Mahvash Jafari
- Department of Biochemistry and Research Center of Chemical Injuries, Faculty of Medicine, Baqiyatallah Medical Sciences University, Tehran, Iran.
| |
Collapse
|
50
|
Widmer R, Ziaja I, Grune T. Protein oxidation and degradation during aging: role in skin aging and neurodegeneration. Free Radic Res 2007; 40:1259-68. [PMID: 17090415 DOI: 10.1080/10715760600911154] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
During aging, the products of oxidative processes accumulate and might disturb cellular metabolism. Among them are oxidized proteins and protein aggregates. On the other hand, in a functioning metabolic system oxidized proteins are degraded, mainly by the proteasome. During aging, however, proteasome activity declines. Therefore, the ability to degrade oxidized proteins is attenuated. The following review summarises the accumulation of oxidized proteins and the decline of the proteasomal system during skin and brain aging including some age-related neurodegenerative processes. The role of protein aggregates will be discussed as a potential reason for the accelerated dysfunction of tissue during aging.
Collapse
Affiliation(s)
- Rebecca Widmer
- Research Institute of Environmental Medicine, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | | | | |
Collapse
|